Language selection

Search

Patent 2546932 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2546932
(54) English Title: QUINAZOLINONE COMPOUNDS AS ANTICANCER AGENTS
(54) French Title: COMPOSES QUINAZOLINONE UTILISES EN TANT QU'AGENTS ANTICANCEREUX
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07D 239/90 (2006.01)
  • A61K 31/505 (2006.01)
  • A61P 35/00 (2006.01)
  • C07D 401/06 (2006.01)
(72) Inventors :
  • WANG, WEIBO (United States of America)
  • LAGNITON, LIANA M. (United States of America)
  • CONSTANTINE, RYAN N. (United States of America)
  • DESAI, MANOJ C. (United States of America)
(73) Owners :
  • NOVARTIS VACCINES AND DIAGNOSTICS, INC. (United States of America)
(71) Applicants :
  • CHIRON CORPORATION (United States of America)
(74) Agent: BORDEN LADNER GERVAIS LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2004-11-24
(87) Open to Public Inspection: 2005-06-09
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2004/039448
(87) International Publication Number: WO2005/051922
(85) National Entry: 2006-05-23

(30) Application Priority Data:
Application No. Country/Territory Date
60/525,059 United States of America 2003-11-25

Abstracts

English Abstract




Quinazolinone compounds, pharmaceutically acceptable salts, and prodrugs
thereof; compositions that include a pharmaceutically acceptable carrier and
one or more of the quinazolinone compounds, either alone or in combination
with at least one additional therapeutic agent. Methods of using the
quinazolinone compounds, either alone or in combination with at least one
additional therapeutic agent as KSP inhibitors, in the prophylaxis or
treatment of proliferative diseases.


French Abstract

L'invention concerne des composés quinazolinone, des sels pharmaceutiquement acceptables et des promédicaments de ces composés, ainsi que des compositions contenant un excipient pharmaceutiquement acceptable et un ou plusieurs desdits composés quinazolinone, seul(s) ou en association avec au moins un agent thérapeutique supplémentaire. La présente invention se rapporte également à des procédés d'utilisation des composés quinazolinone, seuls ou en association avec au moins un agent thérapeutique supplémentaire, en tant qu'inhibiteurs de KSP, pour prévenir ou traiter des maladies impliquant une prolifération.

Claims

Note: Claims are shown in the official language in which they were submitted.



CLAIMS:

The embodiments of the invention in which an exclusive property or privilege
is
claimed are defined as follows:

1. A compound having the formula:
Image
or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug
thereof,
wherein
X is O or S;
R1 is selected from the group consisting of
(1) hydrogen,
(2) substituted or unsubstituted alkyl,
(3) substituted or unsubstituted alkenyl,
(4) substituted or unsubstituted alkynyl,
(5) substituted or unsubstituted aryl,
(6) substituted or unsubstituted heteroaryl,
(7) substituted or unsubstituted heterocyclyl,
(8) substituted or unsubstituted alkylsulfonyl, and
(9) substituted or unsubstituted arylsulfonyl;
R2 is selected from the group consisting of
(1) hydrogen,
(2) substituted or unsubstituted alkyl,
(3) substituted or unsubstituted alkenyl, and
(4) substituted or unsubstituted alkynyl;
R3 is selected from the group consisting of
(1) CO2R10,
(2) COR10,
(3) CONR11R12,



-51-


(4) S(O)m R13, and
(5) SO2NR14R15; or
R2 and R3 taken together with the carbon atom to which they are attached form
a
3- to 7-membered carbocyclic or heterocyclic ring;
with the proviso that when R4 and R5 are taken together to form a 5- to
12-membered heterocyclic ring, R3 is CONR11R12 or R2 and R3 taken together
with the
carbon atom to which they are attached form a 3- to 7-membered carbocyclic or
heterocyclic ring;
R4 is selected from the group consisting of
(1) hydrogen,
(2) substituted or unsubstituted alkyl,
(3) substituted or unsubstituted alkenyl,
(4) substituted or unsubstituted alkynyl,
(5) substituted or unsubstituted aryl,
(6) substituted or unsubstituted heteroaryl, and
(7) substituted or unsubstituted heterocyclyl;
R5 is selected from the group consisting of
(1) hydrogen,
(2) substituted or unsubstituted alkyl,
(3) substituted or unsubstituted alkoxy,
(4) substituted or unsubstituted aryl,
(5) substituted or unsubstituted heteroaryl,
(6) substituted or unsubstituted heterocyclyl,
(7) COR17,
(8) CO2R18,
(9) CONR19R20, and
(10) SO2R21; or
R4 and R5 are taken together with the nitrogen atom to which they are attached
form a heteroaryl or heterocyclyl ring, wherein the heteroaryl ring contains
one or two
ring heteroatoms, wherein the heterocyclyl ring contains one or two ring
heteroatoms, and
wherein the heteroaryl or heterocyclyl ring is optionally substituted with a
halogen, alkyl,
hydroxy, amino, cyano, alkylamino, dialkylamino, alkylaminoalkyl,
dialkylaminoalkyl,
alkoxy, aryl, aryloxy, heteroaryl, arylalkyl, heterocycle, aminocarbonyl,
carbonylamino,



-52-


alkylcarbonyl, alkylcarboxy, alkylaminocarbonyl, alkylcarbonylamino,
carbocycle, or
heteroarylalkyl group; with the proviso that when R4 and R5 taken together
with the
nitrogen atom to which they are attached form a 5-membered heterocyclic ring,
the
heterocyclic ring is not a 2,4-dioxo-3-oxazolidinyl ring, a 2,5-dioxo-1-
imidazolidinyl
ring, or a 2,4,5-trioxo-1-imidazolidinyl ring;
R6, R7, R8, and R9 are independently selected from the group consisting of
(1) hydrogen,
(2) halogen,
(3) hydroxy,
(4) nitro,
(5) amino,
(6) cyano,
(7) alkoxy,
(8) alkylthio,
(9) methylenedioxy,
(10) haloalkoxy,
(11) CO2R10,
(12) COR10,
(13) OR10,
(14) CONR11R12,
(15) substituted or unsubstituted alkyl,
(16) substituted or unsubstituted aryl,
(17) substituted or unsubstituted heteroaryl,
(18) substituted or unsubstituted alkylamino,
(19) substituted or unsubstituted dialkylamino,
(20) substituted or unsubstituted alkylsulfonyl,
(21) substituted or unsubstituted arylsulfonyl,
(22) substituted or unsubstituted alkylcarboxy,
(23) substituted or unsubstituted carboxamido,
(24) substituted or unsubstituted carboxyamino,
(25) substituted or unsubstituted aminocarboxy,
(26) substituted or unsubstituted aminocarbonyl, and
(27) substituted or unsubstituted alkylsulfonamido;



-53-


R10, R11, R12, R13, R14, R15, R17, R18, R19, R20, and R21 are independently
selected from the group consisting of
(1) hydrogen,
(2) substituted or unsubstituted alkyl,
(3) substituted or unsubstituted alkenyl,
(4) substituted or unsubstituted alkynyl,
(5) substituted or unsubstituted aryl,
(6) substituted or unsubstituted heteroaryl, and
(7) substituted or unsubstituted heterocyclyl; or
R11 and R12, R14 and R15, or R19 and R20 taken together form a 3- to
7-membered carbocyclic or heterocyclic ring; and
m = 0, 1, or 2.
2. A compound of Claim 1, wherein substituted alkyl comprises arylalkyl,
heteroarylalkyl, heterocyclyalkyl, aminoalkyl, alkylaminoalkyl,
dialkyaminoalkyl, or
sulfonamidoalkyl.
3. A compound of Claim 1, wherein X is O.
4. A compound of Claim 1, wherein R1 is substituted or unsubstituted
arylalkyl.
5. A compound of Claim 4, wherein R1 is benzyl.
6. A compound of Claim 4, Wherein R1 is substituted benzyl.
7. A compound of Claim 6, wherein R1 is a halo-substituted benzyl.
8. A compound of Claim 7, wherein R1 is 3-chlorobenzyl.
9. A compound of Claim 7, wherein R1 is 3-fluorobenzyl.
10. A compound of Claim 7, wherein R1 is 3-methoxybenzyl.
11. A compound of Claim 7, wherein R1 is 3-trifluoromethylbenzyl.
12. A compound of Claim 7, wherein R1 is 3-trifluoromethoxybenzyl.



-54-


13. A compound of Claim 6, wherein R1 is 3,5-dimethylbenzyl.
14. A compound of Claim 1, wherein R1 is 2-naphthylmethyl.
15. A compound of Claim 1, wherein R2 is hydrogen and R3 is CO2R10.
16. A compound of Claim 15, wherein R10 is alkyl.
17. A compound of Claim 1, wherein R2 is hydrogen and R3 is CONR11R12.
18. A compound of Claim 17, wherein R11 and R12 are alkyl.
19. A compound of Claim 18, wherein R11 and R12 are methyl.
20. A compound of Claim 17, wherein R11 and R12 taken together with the
nitrogen atom to which they are attached from a 3- to 7-membered heterocyclic
ring.
21. A compound of Claim 1, wherein R4 is amino-substituted alkyl.
22. A compound of Claim 21, wherein R4 is 3-aminopropyl.
23. A compound of Claim 1, wherein R5 is hydrogen, alkyl, aryl, or COR17.
24. A compound of Claim 23, wherein R5 is COR17.
25. A compound of Claim 24, wherein R17 is aryl, arylalkyl, alkyl-substituted
aryl, or halogen-substituted aryl.
26. A compound of Claim 25, wherein aryl is phenyl
27. A compound of Claim 1, wherein R6, R8, and R9 are hydrogen.
28. A compound of Claim 1, wherein R7 is a halogen.



-55-


29. A compound of Claim 1 selected from the group consisting of:
ethyl (3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl){(4-bromobenzoyl)[3-
(dimethylamino)propyl]amino}acetate;
ethyl [(3-aminopropyl)(4-methylbenzoyl)amino](3-benzyl-7-chloro-4-oxo-3,4-
dihydroquinazolin-2-yl)acetate;
isopropyl (3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)[[3-
(dimethylamino)propyl](4-methylbenzoyl)amino]acetate;
isopropyl [(3-aminopropyl)(4-bromobenzoyl)amino](3-benzyl-7-chloro-4-oxo-
3,4-dihydroquinazolin-2-yl)acetate;
isopropyl [(3-aminopropyl)(4-methylbenzoyl)amino](3-benzyl-7-chloro-4-oxo-
3,4-dihydroquinazolin-2-yl)acetate;
N-(3-aminopropyl)-N-[1-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
2-(diethylamino)-2-oxoethyl]-4-bromobenzamide;
N-(3-aminopropyl)-N-[1-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
2-(diethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
2-oxo-2-pyrrolidin-1-ylethyl]-4-bromobenzamide;
N-(3-aminopropyl)-N-[1-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
2-oxo-2-pyrrolidin-1-ylethyl]-4-chlorobenzamide;
N-(3-aminopropyl)-N-[1-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
2-morpholin-4-yl-2-oxoethyl]-4-bromobenzamide;
N-(3-aminopropyl)-N-[1-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
2-morpholin-4-yl-2-oxoethyl]-4-chlorobenzamide;
N-(3-aminopropyl)-N-[1-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
2-(dimethylamino)-2-oxoethyl]-4-bromobenzamide;
N-(3-aminopropyl)-N-[1-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
2-(dimethylamino)-2-oxoethyl]-4-chlorobenzamide;
N-(3-aminopropyl)-N-[1-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
2-oxo-2-pyrrolidin-1-ylethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;



-56-




N-(3-aminopropyl)-N-[1-[7-chloro-3-(3-chlorobenzyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-[7-chloro-3-(3-fluorobenzyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-[7-chloro-3-(3-methylbenzyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-{7-chloro-4-oxo-3-[4-(trifluoromethoxy)benzyl]-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-[7-chloro-3-(4-chlorobenzyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-[7-chloro-3-(3-methoxybenzyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylaanino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-[7-chloro-3-(2-naphthylmethyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-{7-chloro-4-oxo-3-[3-(trifluoromethyl)benzyl]-3,4-
dihydroquinazolin-2-yl}-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-[7-chloro-4-oxo-3-[3-(trifluoromethoxy)benzyl]-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-{7-chloro-3-(3-hydroxybenzyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-[7-chloro-3-(3,5-dimethylbenzyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-[3-(1,1'-biphenyl-3-ylmethyl)-7-chloro-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-[7-chloro-3-(3-cyanobenzyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-[7-chloro-3-(3-nitrobenzyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-[7-chloro-3-(3,4-dichlorobenzyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-{1-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-y1)-
2-[ethyl(methyl)amino]-2-oxoethyl)-4-methylbenzamide;
-57-


N-(3-aminopropyl)-N-{-[7-chloro-3-(3,5-dimethylbenzyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-[ethyl(methyl)amino]-2-oxoethyl)-4-methylbenzamide;
N-(3-aminopropyl)-N-{1-[7-chloro-3-(3,5-dimethylbenzyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-[ethyl(methyl)amino]-2-oxoethyl}-4-methylbenzamide;
N-(3-aminopropyl)-N-{1-[7-chloro-3-(3-methoxybenzyl)-4-oxo-3,4
dihydroquinazolin-2-yl]-2-[ethyl(methyl)amino]-2-oxoethyl}-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-[7-chloro-3-(3,5-difluorobenzyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-{7-chloro-3-[(6-chloropyridin-2-yl)methyl]-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide;
N-(3-aminopropyl)-N-[1-{7-chloro-3-[(2-chloropyridin-4-yl)methyl]-4-oxo-3,4-
dihydroquinazolin-2-yl}-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide; and
N-(3-aminopropyl)-N-[1-[7-chloro-4-oxo-3-(quinolin-2-ylmethyl)-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide.

30. A composition, comprising a pharmaceutically acceptable carrier and an
amount of a compound of Claim 1 effective to inhibit KSP activity in a human
or animal
subject when administered thereto.

31. The composition of Claim 30 further comprising at least one additional
agent for the treatment of cancer.

32. The composition of Claim 31, wherein the at least one additional agent for
the treatment of cancer is selected from irinotecan, topotecan, gemcitabine,
gleevec,
herceptin, 5-fluorouracil, leucovorin, carboplatin, cisplatin, taxanes,
tezacitabine,
cyclophosphamide, vinca alkaloids, imatinib, anthracyclines, rituximab, and
trastuzumab.

33. A method for treating a condition by modulation of KSP protein activity
comprising administering to a human or animal subject in need of such
treatment an
effective amount of a compound of Claim 1.

34. The method of Claim 33, wherein the compound has an IC50 value of less
than about 50 µM in a cell proliferation assay.

35. The method of Claim 33, wherein the condition is cancer.
-58-




36. A method for inhibiting KSP activity in a human or animal subject,
comprising administering to the human or animal subject a composition
comprising an
amount of a compound of Claim 1 effective to inhibit KSP activity the human or
animal
subject.
37. A method for treating a cancer disorder in a human or animal subject,
comprising administering to the human or animal subject a composition
comprising an
amount of a compound of Claim 1 effective to inhibit KSP activity the human or
animal
subject.
38. The method of Claim 37 further comprising administering to the human or
animal subject at least one additional agent for the treatment of cancer.
39. The method of Claim 38, wherein the at least one additional agent for the
treatment of cancer is selected from irinotecan, topotecan, gemcitabine,
gleevec,
herceptin, 5-fluorouracil, leucovorin, carboplatin, cisplatin, taxanes,
tezacitabine,
cyclophosphamide, vinca alkaloids, imatinib, anthracyclines, rituximab, and
trastuzumab.
40. A compound of Claim 1 for use in the treatment of cancer.
41. Use of a compound of Claim 1 in the manufacture of a medicament for the
treatment of cancer.
-59-

Description

Note: Descriptions are shown in the official language in which they were submitted.




CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
QUINAZOLINONE COMPOUNDS AS ANTICANCER AGENTS
FIELD OF THE INVENTION
The present invention relates to new quinazolinone compounds, their
pharmaceutically acceptable salts, and prodrugs thereof; compositions of the
new
compounds, either alone or in combination with at least one additional
therapeutic agent,
with a pharmaceutically acceptable carrier; and uses of the new compounds,
either alone
or in combination with at least one additional therapeutic agent, in the
prophylaxis or
treatment of proliferative diseases.
BACKGROUND OF THE INVENTION
Kinesins are motor proteins that use adenosine triphosphate to bind to
microtubules and generate mechanical force. Kinesins are characterized by a
motor
domain having about 350 amino acid residues. The crystal structures of several
kinesin
motor domains have been resolved.
Currently, about one hundred kinesin-related proteins (KRP) have been
identified.
Kinesins are involved in a variety of cell biological processes including
transport of
organelles and vesicles, and maintenance of the endoplasmatic reticulum.
Several KRPs
interact with the microtubules of the mitotic spindle or with the chromosomes
directly,
and appear to play a pivotal role during the mitotic stages of the cell cycle.
These mitotic
KRPs are of particular interest for the development of cancer therapeutics.
Kinesin spindle protein (KSP) (also known as EgS, HsEgS, KNSL1, or KIFII) is
one of several kinesin-like motor proteins that are localized to the mitotic
spindle and
known to be required for formation and/or function of the bipolar mitotic
spindle.
In 1995, the depletion of KSP using an antibody directed against the C-
terminus
of KSP was shown to arrest HeLa cells in mitosis with monoastral microtubule
arrays
(Blangy et al., Cell 83:1159-1169, 1995). Mutations in bimC and cut? genes,
which are
considered to be homologues of KSP, cause failure in centrosome separation in
Aspergillus nidulans (Enos, A.P., and N.R. Morris, Cell 60:1019-1027, 1990)
and
Schizosaccharomyces pombe (Hagan, L, and M. Yanagida, Natuf~e 347:563-566,
1990).
Treatment of cells with either ATRA (all trans-retinoic acid), which reduces
KSP
expression on protein level, or depletion of KSP using antisense
oligonucleotides
_1_



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
revealed a significant growth inhibition in DAN-G pancreatic carcinoma cells
indicating
that KSP might be involved in the antiproliferative action of all traps-
retinoic acid
(Kaiser, A., et al., J. Biol. Chem. 274, 18925-18931, 1999). Interestingly,
the Xenopus
laevis Aurora-related protein kinase pEg2 was shown to associate and
phosphorylate
XlEgS (Diet, R., et al., J. Biol. Chem. 274:15005-15013, 1999). Potential
substrates of
Aurora-related kinases are of particular interest for cancer drug development.
For
example, Aurora l and 2 kinases are overexpressed on protein and RNA level and
the
genes are amplified in colon cancer patients.
The first cell permeable small molecule inhibitor for KSP, "monastrol," was
slaown to arrest cells with monopolar spindles without affecting microtubule
polymerization as do conventional chemotherapeutics such as taxanes and vinca
alkaloids
(Mayer, T.U., et al., Science 286:971-974, 1999). Monastrol was identified as
an
inhibitor in phenotype-based screens and it was suggested that this compound
may serve
as a lead for the development of anticancer drugs. The inhibition was
determined not to
be competitive in respect to adenosine triphosphate and to be rapidly
reversible (DeBonis,
S., et al., Biochemistry 42:338-349, 2003; Kapoor, T.M., et al., J. Cell Biol.
150:975-988,
2000).
Recently, other KSP kinesin inhibitors have been described. WO 02/057244 and
WO 02/056880 describe phenothiazine compounds and triphenylmethane compounds,
respectively, for treating proliferative diseases. WO 02/078639 describes
cyano
substituted dihydropyrimidine compounds for treating proliferative diseases.
U.S. Patent
No. 6,472,521 describes oligonucleotides and oligonucleotide derivatives for
inhibiting
human KSP expression.
WO 01/98278, WO 01/30768, and WO 03/039460 describe quinazolinone
compounds that are useful in treating cellular proliferative diseases
associated with
KSP activity. The compounds described in these references are
2-(2-aminomethyl)quinazolinone derivatives. The quinazolinone compounds
described
in WO 01/98278 and WO 01/30768 have 2-aminomethyl substituents that are either
amine, amide, or sulfonamide substituents. The quinazolinone compounds
described in
WO 03/039460 have the amino group of the 2-aminomethyl substituent
incorporated into
a 5-12 membered nitrogen-containing heterocycle.
-2-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
WO 03/050064 describes thienopyrimidinone compounds that are useful for
treating cellular proliferative disease, for treating disorders associated
with KSP activity,
and for inhibiting KSP.
WO 031103575 describes heterocyclic-fused pyrimidinone derivatives that are
inhibitors of the mitotic KSP and that are useful in the treatment of cellular
proliferative
diseases. These derivatives are N-heterocyclic-fused pyrimidinone derivatives.
Representative derivatives that are described include pyrido[a,(3-y]pyrimidin-
8-ones,
pyrimido[a,(3-y]pyrimidin-~-ones, pyrimido[a,(3-y]pyridazin-8-ones, and
pteridin-4-ones.
SUMMARY OF THE INVENTION
In one aspect of the present invention, new quinazolinone compounds, their
pharmaceutically acceptable salts, and prodrugs thereof axe provided. The
quinazolinone
compounds, pharmaceutically acceptable salts, and prodrugs axe KSP inhibitors
and are
useful in the treating cellular proliferation diseases.
In one embodiment, the quinazolinone compounds have the formula (I):
R9 X
Ra / N/R~
\ ~ / R2
R ~ ~N
R3
6
Rs ~ R4
I
or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug
thereof,
wherein
XisOorS;
Rt is selected from the group consisting of
(1) hydrogen,


(2) substituted or unsubstituted
alkyl,


(3) substituted or unsubstituted
alkenyl,


(4) substituted or unsubstituted
alkynyl,


(5) substituted or unsubstituted
aryl,


(6) substituted or unsubstituted
heteroaryl,


(7) substituted or unsubstituted
heterocyclyl,


-3-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
(8) substituted or unsubstituted alkylsulfonyl, and
(9) substituted or unsubstituted arylsulfonyl;
R2 is selected from the group consisting of
(1) hydrogen,


(2) substituted or unsubstituted
alkyl,


(3) substituted or unsubstituted
alkenyl, and


(4) substituted or unsubstituted
alkynyl;


R3 is selected
from the group
consisting of


(1) C02R10,


(2) CORlo,


(3) CONR11R12~


S(O)mRl3~ and


(5) S02NR14R15~ or


R2 and R3 taken together with the carbon atom to which they are attached form
a
3- to 7-membered carbocyclic or heterocyclic ring;
with the proviso that when R4 and RS are taken together to form a 5- to
12-membered heterocyclic ring, R3 is CONR11R1~ or Ra and R3 taken together
with the
carbon atom to which they are attached form a 3- to 7-membered carbocyclic or
heterocyclic ring;
R4 is selected from the group consisting of
(1) hydrogen,


(2) substituted or unsubstituted
alkyl,


(3) substituted or unsubstituted
alkenyl,


(4) substituted or unsubstituted
alkynyl,


(5) substituted or unsubstituted
aryl,


(6) substituted or unsubstituted
heteroaryl, and


(7) substituted or unsubstituted
heterocyclyl;


RS is selected
from the group
consisting of


(1) hydrogen,


(2) substituted or unsubstituted
alkyl,


(3) substituted or unsubstituted
alkoxy,


(4) substituted or unsubstituted
aryl,


(5) substituted or unsubstituted
heteroaryl,


-4-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
(6) substituted or unsubstituted heterocyclyl,
(7) CORD,
(8) C02R18~
(9) CONR19R2o, and
' (10) S02R21; or
R4 and RS are taken together with the nitrogen atom to which they are attached
form a heteroaryl or heterocyclyl ring, wherein the heteroaryl ring contains
one or two
ring heteroatoms, wherein the heterocyclyl ring contains one or two ring
heteroatoms, and
wherein the heteroaryl or heterocyclyl ring is optionally substituted with a
halogen, alkyl,
hydroxy, amino, cyano, alkylamino, dialkylamino, alkylaminoalkyl,
dialkylaminoalkyl,
alkoxy, aryl, aryloxy, heteroaryl, arylalkyl, heterocycle, aminocarbonyl,
carbonylamino,
alkylcarhonyl, alkylcarboxy, alkylaminocarbonyl, alkylcarbonylamino,
carbocycle, or
heteroarylalkyl group; with the proviso that when R4 and RS taken together
with the
nitrogen atom to which they are attached form a 5-membered heterocyclic ring,
the
heterocyclic ring is not a 2,4-dioxo-3-oxazolidinyl ring, a 2,5-dioxo-1-
imidazolidinyl
ring, or a 2,4,5-trioxo-1-imidazolidinyl ring;
Rg, R~, Rg, and R9 are independently selected from the group consisting of
(1) hydrogen,


(2) halogen,


(3) hydroxy,


(4) nitro,


(5) amino,


(6) cyano,


(7) alkoxy,


(8) alkylthio,


(9) methylenedioxy,


(10) haloalkoxy,


(11) CO~RIO,


(12) CORlo,


(13) ORlo,


(14) CONRIIRt~,


(15) substituted or unsubstituted alkyl,
(16) substituted or unsubstituted aryl,
-5-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
(17) substituted or unsubstituted heteroaryl,


(1~) substituted or unsubstituted alkylamino,


(19) substituted or unsubstituted dialkylamino,


(20) substituted or unsubstituted alkylsulfonyl,


(21) substituted or unsubstituted arylsulfonyl,


(22) substituted or unsubstituted alkylcarboxy,


(23) substituted or unsubstituted carboxamido,


(24) substituted or unsubstituted carboxyamino,


(25) substituted or unsubstituted aminocarboxy,


(26) substituted or unsubstituted aminocarbonyl,
and


(27) substituted or unsubstituted alkylsulfonamido;


Rlo~ Rll~ R12~
R13' R14~ R15~
R17~ R18~ r'19~
R20~ ~d R21 ~e
independently



selected from the group consisting of
(1) hydrogen,
(2) substituted or unsubstituted alkyl,
(3) substituted or unsubstituted alkenyl,
(4) substituted or unsubstituted alkynyl,
(5) substituted or unsubstituted aryl,
(6) substituted or unsubstituted heteroaryl, and
(7) substituted or unsubstituted heterocyclyl; or
Rll and R12, Rlq and Rls, or Rlg and R2p taken together form a 3- to
7-membered carbocyclic or heterocyclic ring; and
m=O, l,or2.
In another aspect, the present invention provides methods for treating
proliferative
diseases in a human or animal subject in need of such treatment comprising
administering
to said subject an amount of a compound of formula (I) effective to reduce or
prevent
cellular proliferation in the subject.
In another aspect of the present invention, methods for treating proliferative
diseases in a human or animal subject in need of such treatment, comprising
administering to said subject an amount of a compound of formula (I) effective
to reduce
or prevent cellular proliferation in the subject in combination with at least
one additional
agent for the treatment of cancer.
-6-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
In other aspects, the present invention provides therapeutic compositions,
comprising at least one compound of formula (I) in combination with one or
more
additional agents for the treatment of cancer, as are commonly employed in
cancer
therapy.
The compounds of the invention are useful in the treatment of cancers,
including,
for example, lung and bronchus; prostate; breast; pancreas; colon and rectum;
thyroid;
stomach; liver and intrahepatic bile duct; kidney and renal pelvis; urinary
bladder; uterine
corpus; uterine cervix; ovary; multiple myeloma; esophagus; acute myelogenous
leukemia; chronic myelogenous leukemia; lymphocytic leukemia; myeloid
leukemia;
brain; oral cavity and pharynx; larynx; small intestine; non-hodgkin lymphoma;
melanoma; and villous colon adenoma.
The invention further provides compositions, kits, methods of use, and methods
of
manufacture as described in the detailed description of the invention.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT
In one aspect of the present invention, new quinazolinone compounds, their
pharmaceutically acceptable salts, and prodrugs thereof are provided. The
quinazolinone
compounds, pharmaceutically acceptable salts, and prodrugs are KSP inhibitors
and are
useful in the treating cellular proliferation diseases.
The quinazolinone compounds have the formula (~:
Ra NiR~
R2
R~ Rs
Rs /Nw
R5 R4
I
or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug
thereof,
wherein
XisOorS;
Rl is selected from the group consisting of
(1) hydrogen,
(2) substituted or unsubstituted alkyl,
(3) substituted or unsubstituted alkenyl,



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
(4) substituted or unsubstituted
alkynyl,


(5) substituted or unsubstituted
aryl,


(6) substituted or unsubstituted
heteroaryl,


(7) substituted or unsubstituted
heterocyclyl,


(8) substituted or unsubstituted
alkylsulfonyl, and


(9) substituted or unsubstituted
arylsulfonyl;


R2 is selected
from the group
consisting of


(1) hydrogen,


(2) substituted or unsubstituted
alkyl,


(3) substituted or unsubstituted
alkenyl, and


(4) substituted or unsubstituted
alkynyl;


R3 is selected
from the group
consisting of


(1) C02R1o,


(2) CORlp,


(3) CONRl1R12~


s(~)mRl3~ and


(5) S02NR14R15~ or


RZ and R3 taken together with the carbon atom to which they are attached form
a
3- to 7-membered carbocyclic or heterocyclic ring;
with the proviso that when R4 and R5 are taken together to form a 5- to
12-membered heterocyclic ring, R3 is CONR11R12 or R2 and R3 taken together
with the
carbon atom to which they are attached form. a 3- to 7-membered carbocyclic or
heterocyclic ring;
R4 is selected from the group consisting of
(1) hydrogen,


(2) substituted or unsubstituted
alkyl,


(3) substituted or unsubstituted
alkenyl,


(4) substituted or unsubstituted
alkynyl,


(5) substituted or unsubstituted
aryl,


(6) substituted or unsubstituted
heteroaryl, and


(7) substituted or unsubstituted
heterocyclyl;


R5 is selected
from the group
consisting of



(1) hydrogen,
_g_



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
(2) substituted or unsubstituted alkyl,
(3) substituted or unsubstituted alkoxy,
(4) substituted or unsubstituted aryl,
(5) substituted or unsubstituted heteroaryl,
(6) substituted or unsubstituted heterocyclyl,
(7) CORD,
(8) C02R1g~
(9) CONRIgR2o, and
(10) SO~R~1; or
R4 and RS are taken together with the nitrogen atom to which they are attached
form a heteroaryl or heterocyclyl ring, wherein the heteroaryl ring contains
one or two
ring heteroatoms, wherein the heterocyclyl ring contains one or two ring
heteroatoms, and
wherein the heteroaryl or heterocyclyl ring is optionally substituted with a
halogen, alkyl,
hydroxy, amino, cyano, alkylamino, dialkylamino, alkylaminoalkyl,
dialkylaminoalkyl,
alkoxy, aryl, aryloxy, heteroaryl, arylalkyl, heterocycle, aminocarbonyl,
carbonylamino,
alkylcarbonyl, alkylcarboxy, alkylaminocarbonyl, alkylcarbonylamino,
carbocycle, or
heteroarylalkyl group; with the proviso that when R4 and RS taken together
with the
nitrogen atom to which they are attached form a 5-membered heterocyclic ring,
the
heterocyclic ring is not a 2,4-dioxo-3-oxazolidinyl ring, a 2,5-dioxo-1-
imidazolidinyl
ring, or a 2,4,5-trioxo-1-imidazolidinyl ring;
R6, R~, Rg, and Rg are independently selected from the group consisting of
(1) hydrogen,
(2) halogen,
(3) hydroxy,
(4) nitro,
(5) amino,
(6) cyano,
(7) alkoxy,
(8) alkylthio,
(9) methylenedioxy,
(10) haloalkoxy,
(11) C02Rlo,
(12) CORlo,
-9-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
(13) OR10,
(14) CONR11R12~
(15) substituted or unsubstituted alkyl,


(16) substituted or unsubstituted aryl,


(17) substituted or unsubstituted heteroaryl,


(1 ~) substituted or unsubstituted alkylamino,


(19) substituted or unsubstituted dialkylamino,


(20) substituted or unsubstituted alkylsulfonyl,


(21) substituted or unsubstituted arylsulfonyl,


(22) substituted or unsubstituted alkylcaxboxy,


(23) substituted or unsubstituted carboxamido,


(24) substituted or unsubstituted carboxyamino,


(25) substituted or unsubstituted aminocaxboxy,


(26) substituted or unsubstituted aminocarbonyl,
and


(27) substituted or unsubstituted alkylsulfonamido;


R10~ Rll~ R12~ R13~ R14~ R15~ R17~ Rls~ R19~ R2o~ and R21 are independently
selected from the group consisting of
(1) hydrogen,
(2) substituted or unsubstituted alkyl,
(3) substituted or unsubstituted alkenyl,
(4) substituted or unsubstituted alkynyl,
(5) substituted or unsubstituted aryl,
(6) substituted or unsubstituted heteroaryl, and
(7) substituted or unsubstituted heterocyclyl; or
Rll and R12, R14 and R15, or R19 and R2o taken' together form a 3- to
7-membered carbocyclic or heterocyclic ring; and
m=0, l,or2.
In one embodiment, X is O.
In one embodiment, Rl is arylalkyl. In one embodiment, the arylalkyl is benzyl
or
substituted benzyl. In one embodiment, Rl is a halo-substituted benzyl. In one
embodiment, Rl is 3-chlorobenzyl. In one embodiment, Rl is 3-fluorobenzyl. In
one
embodiment, Rl is 3-methoxybenzyl. In one embodiment, Rl is 3-
trifluoromethylbenzyl.
-10-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
In one embodiment, Rl is 3-trifluoromethoxybenzyl. In one embodiment, Rl is
3,5-dimethylbenzyl. In one embodiment, Rl is 2-naphthylmethyl.
In one embodiment, R2 is hydrogen and R3 is C02Rlo. In one embodiment, Rlo
is alkyl.
In one embodiment, R2 is hydrogen and R3 is CONRt 1R1~. Tn one embodiment,
Rl 1 and Rl~ are alkyl. In one embodiment, Rl 1 and R12 are methyl. In one
embodiment,
Rl l and R12 are taken together to with the nitrogen atom to which they are
attached from
a 3- to 7-membered heterocyclic ring.
In one embodiment, R4 is amino-substituted alkyl. In one embodiment, R4 is
3-aminopropyl.
In one embodiment, RS is hydrogen, alkyl, aryl, or CORD. In one embodiment,
Rl~ is aryl, arylalkyl, alkyl-substituted aryl, or halogen-substituted aryl.
In one embodiment, R6, Rg, and Rg are hydrogen.
In one embodiment, R~ is a halogen.
For the compounds of formula (I), representative substituted alkyl groups
include
arylalkyl, heteroarylalkyl, heterocyclyalkyl, aminoalkyl, alkylaminoalkyl,
dialkyaminoalkyl, and sulfonamidoalkyl groups. Representative substituted aryl
groups
include sulfonamidoaryl groups. Representative substituted heteroaryl groups
include
alkylheteroaryl groups.
In other aspects, the present invention provides methods for manufacture of
compounds of formula (I). Methods of making representative compounds of the
invention are described in Examples 1 and 2. It is further contemplated that,
in addition
to the compounds of formula (I), intermediates and their corresponding methods
of
syntheses are included within the scope of the invention. Representative
compounds of
the invention are illustrated in Table 1 in Example 3.
In other aspects, the present invention provides compositions that include the
KSP
inhibitors described herein, and methods that utilize the KSP inhibitors
described herein.
In one aspect, the present invention provides pharmaceutical compositions
comprising at least one quinazolinone compound (e.g., a compound of formula
(I))
together With a pharmaceutically acceptable carrier suitable for
administration to a human
or animal subject, either alone or together with other antica~icer agents.
A number of suitable anticancer agents to be used as combination therapeutics
are
contemplated for use in the compositions and methods of the present invention.
Suitable
-11-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
anticancer agents to be used in combination with the compounds of the
invention include
agents that induce apoptosis; polynucleotides (e.g., ribozymes); polypeptides
(e.g.,
enzymes); drugs; biological mimetics; alkaloids; alkylating agents; antitumor
antibiotics;
antimetabolites; hormones; platinum compounds; monoclonal antibodies
conjugated with
anticancer drugs, toxins, andlor radionuclides; biological response modifiers
(e.g.,
interferons [e.g., IFN-a] and interleukins [e.g., IL-2]); adoptive
immunotherapy agents;
hematopoietic growth factors; agents that induce tumor cell differentiation
(e.g., all-trans-
retinoic acid); gene therapy reagents; antisense therapy reagents and
nucleotides; tumor
vaccines; inhibitors of angiogenesis, and the like. Numerous other examples of
chemotherapeutic compounds and anticancer therapies suitable for
coadministration with
the disclosed compounds of formula (I) are known to those skilled in the art.
In certain embodiments, anticancer agents to be used in combination with
compounds of the present invention comprise agents that induce or stimulate
apoptosis.
Agents that induce apoptosis include, but are not limited to, radiation;
kinase inhibitors
(e.g., Epidermal Growth Factor Receptor [EGFR] kinase inhibitor, Vascular
Growth
Factor Receptor [VGFR] kinase inhibitor, Fibroblast Growth Factor Receptor
[FGFR]
kinase inhibitor, Platelet-derived Growth Factor Receptor [PGFR] I kinase
inhibitor, and
Bcr-Abl kinase inhibitors such as STI-571, Gleevec, and Glivec]); antisense
molecules;
antibodies [e.g., Herceptin and Rituxan]; anti-estrogens [e.g., raloxifene and
tamoxifen];
anti-androgens [e.g., flutamide, bicalutamide, finasteride, amino-
glutethamide,
ketoconazole, and corticosteroids]; cyclooxygenase 2 (COX-2) inhibitors [e.g.,
Celecoxib, meloxicam, NS-39~, and non-steroidal anti-inflammatory drugs
(NSAIDs)];
and cancer chemotherapeutic drugs [e.g., irinotecan (Camptosar), CPT-11,
fludarabine
(Fludara), dacarbazine (DTIC), dexamethasone, mitoxantrone, Mylotarg, VP-16,
cisplatinum, 5-FU, Doxrubicin, TAXOTERE or TAXOL]; cellular signaling
molecules;
ceramides and cytokines; and staurosprine; and the like.
In other aspects, the invention provides methods for using the compounds
described herein. For example, the compounds described herein can be used in
the
treatment of cancer. The compounds described herein can also be used in the
manufacture of a medicament for the treatment of cancer.
In one embodiment, the present invention provides methods of treating human or
animal subjects suffering from a cellular proliferative disease, such as
cancer. The
present invention provides methods of treating a human or animal subj ect in
need of such
-12-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
treatment, comprising administering to the subject a therapeutically effective
amount of
an quinazolinone compound (e.g., a compound of formula (I)), either alone or
in
combination with other anticancer agents.
In another embodiment, the present invention provides methods for treating a
cellular proliferative disease in a human or animal subject in need of such
treatment
comprising, administering to said subject an amount of an quinazolinone
compound (e.g.,
a compound of formula (I)) effective to reduce or prevent cellular
proliferation or tumor
growth in the subject.
In another embodiment, the present invention provides methods for treating a
cellular proliferative disease in a human or animal subject in need of such
treatment
comprising administering to said subject an amount of an quinazolinone
compound (e.g.,
a compound of formula (1)) effective to reduce or prevent cellular
proliferation in the
subject in combination with at least one additional agent for the treatment of
cancer.
The present invention provides compounds that are inhibitors of KSP. The
inhibitors are useful in pharmaceutical compositions for human or veterinary
use where
inhibition of KSP is indicated, e.g., in the treatment of cellular
proliferative diseases such
as tumor and/or cancerous cell growth mediated by KSP. In particular, the
compounds
are useful in the treatment of human or animal (e.g., murine) cancers,
including, for
example, lung and bronchus; prostate; breast; pancreas; colon and rectum;
thyroid;
stomach; liver and intrahepatic bile duct; kidney and renal pelvis; urinary
bladder; uterine
corpus; uterine cervix; ovary; multiple myeloma; esophagus; acute myelogenous
leukemia; chronic myelogenous leukemia; lymphocytic leukemia; myeloid
leukemia;
brain; oral cavity and pharynx; larynx; small intestine; non-hodgkin lymphoma;
melanoma; and vinous colon adenoma.
In another embodiment, the invention provides methods of treating an KSP
mediated disorder. In one method, an effective amount of an quinazolinone
compound is
administered to a patient (e.g., a human or animal subject) in need thereof to
mediate
(or modulate) KSP activity.
A representative assay for determining KSP inhibitory activity is described in
Example 4.
The following definitions are provided to better understand the invention.
As used herein, the term "quinazolinone" refers to a quinazolinone compound
having a carbonyl or thiocarbonyl group at position 4.
-13-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
"Alkyl" refers to alkyl groups that do not contain heteroatoms. Thus the
phrase
includes straight chain alkyl groups such as methyl, ethyl, propyl, butyl,
pentyl, hexyl,
heptyl, octyl, nonyl, decyl, undecyl, dodecyl and the like. The phrase also
includes
branched chain isomers of straight chain alkyl groups, including but not
limited to, the
following which are provided by way of example: -CH(CH3)z, -CH(CH3)(CH2CH3),
-CH(CHzCH3)z, -C(CH3)3, -C(CH2CH3)3, -CH2CH(CH3)z, -CH2CH(CH3)(CHaCH3)a
-CH2CH(CHZCH3)z, -CHaC(CH3)3, -CH2C(CHZCH3)3, -CH(CH3)-CH(CH3)(CH2CH3),
( -CH2CH2CH(CH3)(CH2CH3), -CHZCH2CH(CH2CH3)z,
-CH2CH2CH CH3)z,
-CH2CHzC(CH3)3, -CH2CHzC(CHzCH3)3, -CH(CH3)CHz_CH(CH3)z,
-CH(CH3)CH(CH3)CH(CH3)z, -CH(CHZCH3)CH(CH3)CH(CH3)(CH2CH3), and others.
The phrase also includes cyclic alkyl groups such as cyclopropyl, cyclobutyl,
cyclopentyl,
cyclohexyl, cycloheptyl, and cyclooctyl and such rings substituted with
straight and
branched chain alkyl groups as defined above. Thus the phrase "alkyl groups"
includes
primary alkyl groups, secondary alkyl groups, and tertiary alkyl groups.
Preferred alkyl
groups include straight and branched chain alkyl groups and cyclic alkyl
groups having
1 to 12 carbon atoms.
"Alkylene" refers to the same residues as noted above for "alkyl," but having
two
points of attachment. Exemplary alkylene groups include ethylene (-CHzCHz-),
propylene (-CH2CH2CHz-), dimethylpropylene (-CHzC(CH3)zCHz-), and
cyclohexylpropylene (-CH2CHzCH(C6H13)-)
"Alkenyl" refers to straight chain, branched, or cyclic radicals having one or
more
carbon-carbon double bonds and from 2 to about 20 carbon atoms. Preferred
alkenyl
groups include straight chain and branched alkenyl groups and cyclic alkenyl
groups
having 2 to 12 carbon atoms.
"Alkynyl" refers to straight chain, branched, or cyclic radicals having one or
more
carbon-carbon triple bonds and from 2 to about 20 carbon atoms. Preferred
alkynyl
groups include straight chain and branched alkynyl groups having 2 to 12
carbon atoms.
Alkyl, alkenyl, and alkynyl groups may be substituted. "Substituted alkyl"
refers
to an allcyl group as defined above in which one or more bonds to a carbons)
or
hydrogen(s) are replaced by a bond to non-hydrogen and non-carbon atoms such
as, but
not limited to, a halogen atom such as F, Cl, Br, and I; an oxygen atom in
groups such as
hydroxyl groups, alkoxy groups, aryloxy groups, and ester groups; a sulfur
atom in
groups such as thiol groups, alkyl and aryl sulfide groups, sulfone groups,
sulfonyl
-14-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
groups, and sulfoxide groups; a nitrogen atom in groups such as amines,
amides,
alkylamines, dialkylaxnines, arylamines, alkylarylamines, diaxylamines, N-
oxides, imides,
and enamines; a silicon atom in groups such as in trialkylsilyl groups,
dialkylarylsilyl
groups, alkyldiarylsilyl groups, and triarylsilyl groups; and other
heteroatoms in various
other groups. Substituted alkyl groups also include groups in which one or
more bonds to
a carbons) or hydrogen(s) atom is replaced by a higher-order bond (e.g., a
double- or
triple-bond) to a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and
ester groups;
nitrogen in groups such as imines, oximes, hydrazones, and nitrites.
Substituted alkyl
groups further include alkyl groups in which one or more bonds to a carbons)
or
hydrogen(s) atoms is replaced by a bond to an aryl, heteroaxyl, heterocyclyl,
or cycloalkyl
group. Preferred substituted alkyl groups include, among others, alkyl groups
in which
one or more bonds to a carbon or hydrogen atom is/are replaced by one or more
bonds to
fluoro, chloro, or bromo group. Another preferred substituted alkyl group is
the
trifluoromethyl group and other alkyl groups that contain the trifluoromethyl
group.
Other preferred substituted alkyl groups include those in which one or more
bonds to a
carbon or hydrogen atom is replaced by a bond to an oxygen atom such that the
substituted alkyl group contains a hydroxyl, alkoxy, or aryloxy group. Other
preferred
substituted alkyl groups include alkyl groups that have an amine, or a
substituted or
unsubstituted alkylamine, dialkylamine, arylamine, (alkyl)(aryl)amine,
diarylamine,
heterocyclylamine, diheterocyclylamine, (alkyl)(heterocyclyl)amine, or
(aryl)(heterocyclyl)amine group. Still other preferred substituted alkyl
groups include
those in which one or more bonds to a carbons) or hydrogen(s) atoms is
replaced by a
bond to an aryl, heteroaryl, heterocyclyl, or cycloalkyl group. Examples of
substituted
alkyl are: -(CHz)3NHz, -(CHz)3NH(CH3), -(CHz)3NH(CH3)z, -CH2C(=CHz)CHzNHz,
_CHZC(=O)CHzNHz,-CHZS(=O)zCH3,-CH20CHzNHz, -C02H. Examples of substituents
of substituted alkyl are: -CH3, -CaHs, -CH20H, -OH, -OCH3, -OC2H5, -OCF3,
-OC(=O)CH3, -OC(=O)NHz, -OC(=O)N(CH3)z, -CN, NOz, -C(=O)CH3, -C02H,
-C02CH3, -CONHz, NHz~ N(CH3)z, -NHS02CH3, NHCOCH3, NHC(=O)OCH3,
-NHSO-zCH3, -S02CH3, -SOzNHz, Halo.
"Substituted alkenyl" has the same meaning with respect to alkenyl groups that
substituted alkyl groups had with respect to unsubstituted alkyl groups. A
substituted
alkenyl group includes alkenyl groups in which a non-carbon or non-hydrogen
atom is
bonded to a carbon double bonded to another carbon and those in which one of
the
-15-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
non-carbon or non-hydrogen atoms is bonded to a carbon not involved in a
double bond
to another carbon.
"Substituted alkynyl" has the same meaning with respect to alkynyl groups that
substituted alkyl groups had with respect to unsubstituted alkyl groups. A
substituted
alkynyl group includes alkynyl groups in which a non-carbon or non-hydrogen
atom is
bonded to a carbon triple bonded to another carbon and those in which a non-
carbon or
non-hydrogen atom is bonded to a carbon not involved in a triple bond to
another carbon.
"Alkoxy" refers to RO- wherein R is alkyl. Representative examples of alkoxy
groups include methoxy, ethoxy, t-butoxy, trifluoromethoxy, and the like.
"Halogen" or "halo" refers to chloro, bromo, fluoro, and iodo groups. The term
"haloalkyl" refers to an alkyl radical substituted with one or more halogen
atoms. The
term "haloalkoxy" refers to an alkoxy radical substituted with one or more
halogen atoms.
"Amino" refers hexein to the group NH2. The term "alkylamino" refers herein to
the group NRR' where R is alkyl and R' is hydrogen or alkyl. The term
"arylamino"
refers herein to the group NRR' where R is aryl and R' is hydrogen, alkyl, or
aryl. The
term "aralkylamino" refers herein to the group NRR' where R is aralkyl and R'
is
hydrogen, alkyl, aryl, or aralkyl.
"Alkoxyallcyl" refers to the group -alkl-O-alk2 where alkl is alkyl or
alkenyl, and
alka is alkyl or allcenyl. The term "aryloxyalkyl" refers to the group -alkyl
O-aryl. The
term "aralkoxyalkyl" refers to the group -alkylenyl-O-aralkyl.
"Alkoxyallcylamino" refers herein to the group NR-( alkoxyalkyl), where R is
typically hydrogen, aralkyl, or alkyl.
"Aminocaxbonyl" refers herein to the group -C(O)-NH2 . "Substituted
aminocarbonyl" refers herein to the group -C(O)-NRR' where R is alkyl and R'
is
hydrogen or alkyl. The term "arylaminocarbonyl" refers herein to the group -
C(O)-NRR'
where R is aryl and R' is hydrogen, alkyl or aryl. "Aralkylarninocarbonyl"
refers herein
to the group -C(O)-NRR' where R is aralkyl and R' is hydrogen, alkyl, aryl, or
aralkyl.
"Aminosulfonyl" refers herein to the group -S(O)2-NHa. "Substituted
aminosulfonyl" refers herein to the group -S(O)2-NRR' where R is alkyl and R'
is
hydrogen or alkyl. The term "aralkylaminosulfonlyaryl" refers herein to the
group
-aryl-S(O)z-NH-aralkyl.
"Carbonyl" refers to the divalent group -C(O)-.
-16-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
"Carbonyloxy" refers generally to the group -C(O)-O. Such groups include
esters, -C(O)-O-R, where R is alkyl, cycloalkyl, aryl, or arallcyl. The term
"carbonyloxycycloalkyl" refers generally herein to both a
"carbonyloxycarbocycloalkyl"
and a "carbonyloxyheterocycloalkyl," i.e., where R is a carbocycloalkyl or
heterocycloalkyl, respectively. The term "arylcarbonyloxy" refers herein to
the group
-C(O)-O-aryl, where aryl is a mono- or polycyclic, carbocycloaryl or
heterocycloaryl.
The term "aralkylcarbonyloxy" refers herein to the group -C(O)-O-aralkyl.
"Sulfonyl" refers herein to the group -SOz-. "Alkylsulfonyl" refers to a
substituted sulfonyl of the structure -S02R- in which R is alkyl.
Alkylsulfonyl groups
employed in compounds of the present invention are typically alkylsulfonyl
groups
having from 1 to 6 carbon atoms in its backbone structure. Thus, typical
alkylsulfonyl
groups employed in compounds of the present invention include, for example,
methylsulfonyl (i.e., where R is methyl), ethylsulfonyl (i.e., where R is
ethyl),
propylsulfonyl (i.e., where R is propyl), and the like. The term
"arylsulfonyl" refers
herein to the group -SOz-aryl. The term "aralkylsulfonyl" refers herein to the
group
-SOz-aralkyl. The term "sulfonamido" refers herein to -SOzNHz.
"Carbonylamino" refers to the divalent group -NH-C(O)- in which the hydrogen
atom of the amide nitrogen of the carbonylamino group can be replaced alkyl,
aryl, or
aralkyl group. Such groups include moieties such as carbamate esters (-NH-C(O)-
O-R)
and amides NH-C(O)-R, where R is a straight or branched chain alkyl,
cycloalkyl, or
aryl or axalkyl. The term "alkylcarbonylamino" refers to alkylcarbonylamino
where R is
alkyl having from 1 to about 6 carbon atoms in its backbone structure. The
term
"arylcarbonylamino" refers to group NH-C(O)-R where R is an aryl. Similarly,
the term
"aralkylcarbonylamino " refers to carbonylamino where R is aralkyl.
"Guanidino" or "guanidyl" refers to moieties derived from guanidine,
HZN-C(=NH)-NHz. Such moieties include those bonded at the nitrogen atom
carrying the
formal double bond (the "2"-position of the guanidine, e.g.,
diaminomethyleneamino,
(H2I~zC=NH-)) and those bonded at either of the nitrogen atoms carrying a
formal single
bond (the "1-" and/or "3"-positions of the guandine, e.g., H2N-C(--NH)-NH-)).
The
hydrogen atoms at any of the nitrogens can be replaced with a suitable
substituent, such
as alkyl, aryl, or aralkyl.
-17-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
"Amidino" refers to the moieties R-C(--N)-NR'- (the radical being at the
"Nl" nitrogen) and R(NR')C--N- (the radical being at the "N2" nitrogen), where
R and R'
can be hydrogen, alkyl, aryl, or aralkyl.
"Cycloalkyl" refers to a mono- or polycyclic, heterocyclic or carbocyclic
alkyl
substituent. Typical cycloalkyl substituents have from 3 to 8 backbone (i.e.,
ring) atoms
in which each backbone atom is either carbon or a heteroatom. The term
"heterocycloalkyl" refers herein to cycloalkyl substituents that have from 1
to 5, and more
typically from 1 to 4 heteroatoms in the ring structure. Suitable heteroatoms
employed in
compounds of the present invention are nitrogen, oxygen, and sulfur.
Representative
heterocycloalkyl moieties include, for example, morpholino, piperazinyl,
piperadinyl, and
the like. Carbocycloalkyl groups are cycloalkyl groups in which all ring atoms
are
carbon. When used in connection with cycloalkyl substituents, the term
"polycyclic"
refers herein to fused and non-fused alkyl cyclic structures.
"Substituted heterocycle," "heterocyclic group," "heterocycle," or
"heterocyclyl,"
as used herein refers to any 3- or 4-membered ring containing a heteroatom
selected from
nitrogen, oxygen, and sulfur or a 5- or 6-membered ring containing from one to
three
heteroatoms selected from the group consisting of nitrogen, oxygen, or sulfur;
wherein
the 5-membered ring has 0-2 double bonds and the 6-membered ring has 0-3
double
bonds; wherein the nitrogen and sulfur atom maybe optionally oxidized; wherein
the
nitrogen and sulfur heteroatoms maybe optionally quarternized; and including
any
bicyclic group in which any of the above heterocyclic rings is fused to a
benzene ring or
another 5- or 6-membered heterocyclic ring independently defined above. The
term
"heterocycle" thus includes rings in which nitrogen is the heteroatom as well
as partially
and fully-saturated rings. Preferred heterocycles include, for example:
diazapinyl, pyrryl,
pyrrolinyl, pyrrolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, imidazoyl,
imidazolinyl,
imidazolidinyl, pyridyl, piperidinyl, pyrazinyl, piperazinyl, N-methyl piper-
azinyl,
azetidinyl, N-methylazetidinyl, pyrimidinyl, pyridazinyl, oxazolyl,
oxazolidinyl,
isoxazolyl, isoazolidinyl, morpholinyl, thiazolyl, thiazolidinyl,
isothiazolyl,
isothiazolidinyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl,
benzothiazolyl,
benzoxazolyl, furyl, thienyl, triazolyl, and benzothienyl.
Heterocyclic moieties can be unsubstituted or monosubstituted or disubstituted
with various substituents independently selected from hydroxy, halo, oxo
(C=O),
-18-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
alkylimino (RN=, wherein R is alkyl or alkoxy group), amino, alkylamino,
dialkylamino,
acylaminoalkyl, alkoxy, thioalkoxy, polyalkoxy, alkyl, cycloalkyl or
haloalkyl.
The heterocyclic groups may be attached at various positions as will be
apparent
to those having skill in the organic and medicinal chemistry arts in
conjunction with the
disclosure herein.
0 0
N ~,
wN wN wN wN~ wN~
~° ~N,
o. °
~N
~N~
~NH
O
wN~ O wN~ O
~N O ~N~ O~N O ~N NHZ
O~NH
N O \N~NH ~N
N ~ 1~ ~o _
°%~ ° ° \ / s
OH R
~N R,N~R
O R~Nw
iN
where R is H or a heterocyclic substituent, as described herein.
Representative heterocyclics include, for example, imidazolyl, pyridyl,
piperazinyl, azetidinyl, tluazolyl, furanyl, ~ triazolyl benzimidazolyl,
benzothiazolyl,
benzoxazolyl, quinolinyl, isoquinolinyl, quinazolinyl, quinoxalinyl,
phthalazinyl, indolyl,
naphthpyridinyl, indazolyl, and quinolizinyl.
"Aryl" refers to optionally substituted monocyclic and polycyclic aromatic
groups
having from 3 to 14 backbone carbon or hetero atoms, and includes both
carbocyclic aryl
groups and heterocyclic aryl groups. Carbocyclic aryl groups are aryl groups
in which all
ring atoms in the aromatic ring are carbon. The term "heteroaryl" refers
herein to aryl
groups having from 1 to 4 heteroatoms as ring atoms in an aromatic ring with
the
remainder of the ring atoms being carbon atoms. When used in connection with
aryl
substituents, the term "polycyclic aryl" refers herein to fused and non-fused
cyclic
structures in which at least one cyclic structure is aromatic, such as, for
example,
-19-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
benzodioxozolo (which has a heterocyclic structure fused to a phenyl group,
i.e.,
°~ , naphthyl, and the like. Exemplary aryl moieties employed as
substituents in
compounds of the present invention include phenyl, pyridyl, pyrimidinyl,
thiazolyl,
indolyl, imidazolyl, oxadiazolyl, tetrazolyl, pyrazinyl, triazolyl,
thiophenyl, furanyl,
quinolinyl, purinyl, naphthyl, benzothiazolyl, benzopyridyl, and
benzimidazolyl, and the
like.
"Aralkyl" or "arylalkyl" refers to an alkyl group substituted with an aryl
group.
Typically, aralkyl groups employed in compounds of the present invention have
from 1 to
6 carbon atoms incorporated within the alkyl portion of the aralkyl group.
Suitable
aralkyl groups employed in compounds of the present invention include, for
example,
benzyl, picolyl, and the like.
Representative heteroaryl groups include, for example, those shown below.
These
heteroaryl groups can be further substituted and may be attached at various
positions as
will be apparent to those having skill in the organic and medicinal chemistry
arts in
conjunction with the disclosure herein.
w i~ ~N y,i
N~ ~N ~ N N
N HN
.~ ~ O O
F F \N- \\ ~-- 'N_N ~N
~N \ F ~ N I N F' I N.N HN /
/N~ F F
O
\N-~
N
N \ \ \
/ ~ \N-N y ~~NH2 N N 'NN
w wN~-' ~ wN'
~O
N eN \
11 0 \N~N N N
N sN N w \
HN.NoN ~ /N~ 1
\
-N
\N~NH N oN
O
\ / ~ /
-20-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Representative heteroaryls include, for example, imidazolyl, pyridyl,
thiazolyl,
triazolyl benzimidazolyl, benzothiazolyl, and benzoxazolyl.
"Biaxyl" refers to a group or substituent to which two aryl groups, which are
not
condensed to each other, are bound. Exemplary biaryl compounds include, for
example,
phenylbenzene, diphenyldiazene, 4-methylthio-1-phenylbenzene, phenoxybenzene,
(2-phenylethynyl)benzene, diphenyl ketone, (4-phenylbuta-1,3-diynyl)benzene,
phenylbenzylamine, (phenylmethoxy)benzene, and the like. Preferred optionally
substituted biaryl groups include: 2-(phenylamino)-N-[4-(2-phenylethynyl)-
phenyl]acetamide, 1,4-diphenylbenzene, N-[4-(2-phenylethynyl)phenyl]-2-[benzyl-

amino]-acetamide, 2-amino-N-[4-(2-phenylethynyl)phenyl]propanamide, 2-amino-N-
[4-
(2-phenyl-ethynyl)phenyl]acetamide, 2-(cyclopropylamino)-N-[4-(2-
phenylethynyl)-
phenyl]-acetamide, 2-(ethylamino)-N-[4-(2-phenylethynyl)phenyl] acetamide,
2-[(2-methyl-propyl)amino]-N-[4-(2-phenylethynyl)phenyl]acetamide, 5-phenyl-2H-

benzo-[d]1,3-dioxolene, 2-chloro-1-methoxy-4-phenylbenzene, 2-
[(imidazolylmethyl)-
amino]-N-[4-(2-phenylethynyl)phenyl]acetamide, 4-phenyl-1-phenoxybenzene,
N-(2-amino-ethyl)-[4-(2-phenylethynyl)phenyl] caxboxamide, 2- { [ (4-
fluorophenyl)methyl]-amino-N-[4-(2-phenylethynyl)phenyl]acetamide, 2-{[(4-
methylphenyl)methyl]amino)-N-[4-(2-phenyl-ethynyl)phenyl]acetamide, 4-phenyl-1-

(trifluoromethyl)benzene, 1-butyl-4-phenyl-benzene, 2-(cyclohexylarnino)-N-[4-
(2-
phenylethynyl)phenyl]acetamide, 2-(ethyl-methyl-amino)-N-[4-(2-
phenylethynyl)phenyl] acetamide, 2-(butylamino)-N-[4-(2-phenyl-ethynyl)-
phenyl]acetamide, N-[4-(2-phenylethynyl)phenyl]-2-(4-pyridylamino)-acetamide,
N-[4-
(2-phenylethynyl)phenyl]-2-(quinuclidin-3-ylamino)acetamide, N-[4-(2-phenyl-
ethynyl)phenyl]pyrrolidin-2-ylcarboxamide, 2-amino-3-methyl-N-[4-(2-phenyl-
ethynyl)-
phenyl]butanamide, 4-(4-phenylbuta-1,3-diynyl)phenylamine, 2-(dimethyl-amino)-
N-[4-
(4-phenylbuta-1,3-diynyl)phenyl]acetamide, 2-(ethylamino)-N-[4-(4-phenylbuta-
1,3-
diynyl)-phenyl]acetamide, 4-ethyl-1-phenylbenzene, 1-[4-(2-phenyl-ethynyl)-
phenyl]ethan-1-one, N-(1-carbamoyl-2-hydroxypropyl)[4-(4-phenylbuta-1,3-
diynyl)-
phenyl]-carbox-amide, N-[4-(2-phenylethynyl)phenyl]propanamide, 4-methoxy-
phenyl
phenyl ketone, phenyl-N-benzamide, (tert-butoxy)-N-[(4-phenylphenyl)-methyl]-
carboxamide, 2-(3-phenyl-phenoxy)ethanehydroxamic acid, 3-phenylphenyl
propanoate,
1-(4-ethoxyphenyl)-4-methoxybenzene, and [~.-(2-phenylethynyl)phenyl]pyrrole.
-21-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
"Heteroarylaryl" refers to a biaryl group where one of the aryl groups is a
heteroaryl group. Exemplary heteroarylaryl groups include, for example,
2-phenylpyridine, phenylpyrrole, 3-(2-phenylethynyl)pyridine, phenylpyrazole,
5-(2-phenyl-ethynyl)-1,3-dihydropyrimidine-2,4-dione, 4-phenyl-1,2,3-
thiadiazole,
2-(2phenyl-ethynyl)pyrazine, 2-phenylthiophene, phenylimidazole, 3-(2-
piperazinyl-
phenyl)-furan, 3-(2,4-dichlorophenyl)-4-methylpyrrole, and the like. Preferred
optionally
substituted heteroarylaryl groups include: 5-(2-phenylethynyl)pyrimidine-2-
ylamine,
1-methoxy-4-(2-thienyl)benzene, 1-methoxy-3-(2-thienyl)benzene, 5-methyl-2-
phenyl-
pyridine, 5-methyl-3-phenylisoxazole, 2-[3-(trifluoromethyl)phenyl]furan, 3-
fluoro-5-
(2-furyl)-2-methoxy-1-prop-2-enylbenzene, (hydroxyimino)(5-phenyl(2-thienyl))-
methane, 5-[(4-methylpiperazinyl)methyl]-2-phenylthiophene, 2-(4-ethylphenyl)-
thio-
phene, 4-methyl-thin-1-(2-thienyl)benzene, 2-(3-nitrophenyl)thiophene, (tert-
butoxy)-N-
[(5-phenyl-(3-pyridyl))methyl]carboxamide, hydroxy-N-[(5-phenyl(3-
pyridyl))methyl]-
amide, 2-(phenyl-methylthio)pyridine, and benzylimidazole.
"Heteroarylheteroaryl" refers to a biaryl group where both of the aryl groups
is a
heteroaryl group. Exemplary heteroarylheteroaryl groups include, for example,
3-pyridylimidazole, 2-imidazolylpyrazine, and the like. Preferred optionally
substituted
heteroarylheteroaryl groups include: 2-(4-piperazinyl-3-pyridyl)fitran,
diethyl-(3-pyrazin
2-yl(4-pyridyl))amine, and dimethyl f 2-[2-(5-methylpyrazin-2-yl)ethynyl](4
pyridyl)~amine.
"Optionally substituted" or "substituted" refers to the replacement of
hydrogen
with a monovalent or divalent radical. Suitable substitution groups include,
for example,
hydroxyl, nitro, amino, imino, cyano, halo, thio, sulfonyl, thioamido,
amidino, imidino,
oxo, oxamidino, methoxamidino, imidino, guanidino, sulfonamido, carboxyl,
formyl,
alkyl, haloalkyl, alkyamino, haloalkylamino, alkoxy, haloalkoxy, alkoxy-alkyl,
alkylcarhonyl, aminocarbonyl, arylcarbonyl, aralkylcarbonyl,
heteroarylcarbonyl,
heteroaralkyl-carbonyl, alkylthio, aminoalkyl, cyanoalkyl, aryl and the like.
The substitution group can itself be substituted. The group substituted onto
the
substitution group can be carboxyl, halo, nitro, amino, cyano, hydroxyl,
alkyl, alkoxy,
aminocarbonyl, -SR, thioamido, -S03H, -SOZR, or cycloalkyl, where R is
typically
hydrogen, hydroxyl or alkyl.
When the substituted substituent includes a straight chain group, the
substitution
can occur either within the chain (e.g., 2-hydroxypropyl, 2-aminobutyl, and
the like) or at
-22-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
the chain terminus (e.g., 2-hydroxyethyl, 3-cyanopropyl, and the like).
Substituted
substituents can be straight chain, branched or cyclic arrangements of
covalently bonded
carbon or heteroatoms.
"Carboxy-protecting group" refers to a carbonyl group which has been
esterified
with one of the commonly used carboxylic acid protecting ester groups employed
to
block or protect the carboxylic acid function while reactions involving other
functional
sites of the compound are carried out. In addition, a carboxy protecting group
can be
attached to a solid support whereby the compound remains connected to the
solid support
as the carboxylate until cleaved by hydrolytic methods to release the
corresponding free
acid. Representative carboxy-protecting groups include, for example, alkyl
esters,
secondary amides and the like.
Certain of the compounds of the invention comprise asymmetrically substituted
carbon atoms. Such asymmetrically substituted carbon atoms can result in the
compounds of the invention comprising mixtures of stereoisomers at a
particular
asymmetrically substituted carbon atom or a single stereoisomer. As a result,
racemic
mixtures, mixtures of diastereomers, as well as single diastereomers of the
compounds of
the invention are included in the present invention. The terms "S" and "R"
configuration,
as used herein, are as defined by the IUPAC 1974 "RECOMMENDATIONS FOR SECTION
E,
FUNDAMENTAL STEREOCHEMISTRY," Pure Appl. Chem. 45:13-30, 1976. The terms oc
and
(3 are employed for ring positions of cyclic compounds. The a-side of the
reference plane ,
is that side on which the preferred substituent lies at the lower numbered
position. Those
substituents lying on the opposite side of the reference plane are assigned (3
descriptor. It
should be noted that this usage differs from that for cyclic stereoparents, in
which "a"
means "below the plane" and denotes absolute configuration. The terms a and (3
configuration, as used herein, are as defined by the "Chemical Abstracts Index
Guide,"
Appendix IV, paragraph 203, 197.
As used herein, the term "pharmaceutically acceptable salts" refers to the
nontoxic
acid or alkaline earth metal salts of the compounds of formula (I). These
salts can be
prepared ira situ during the final isolation and purification of the compounds
of
formula (I), or by separately reacting the base or acid functions with a
suitable organic or
inorganic acid or base, respectively. Representative salts include, but are
not limited to,
the following: acetate, adipate, alginate, citrate, aspartate, benzoate,
benzenesulfonate,
bisulfate, butyrate, camphorate, camphorsulfonate, digluconate,
cyclopentanepropionate,
-23-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
dodecylsulfate, ethanesulfonate, glucoheptanoate, glycerophosphate, hemi-
sulfate,
heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide,
2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, nicotinate, 2-
napth-
alenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-phenylproionate,
picrate,
pivalate, propionate, succinate, sulfate, tartrate, thiocyanate, p-
toluenesulfonate, and
undecanoate. Also, the basic nitrogen-containing groups can be quaternized
with such
agents as alkyl halides, such as methyl, ethyl, propyl, and butyl chloride,
bromides, and
iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl
sulfates, long chain
halides such as decyl, lauryl, myristyl, and stearyl chlorides, bromides and
iodides,
aralkyl halides like benzyl and phenethyl bromides, and others. Water or oil-
soluble or
dispersible products are thereby obtained.
Examples of acids that may be employed to form pharmaceutically acceptable
acid addition salts include. such inorganic acids as hydrochloric acid,
sulfuric acid and
phosphoric acid and such organic acids as oxalic acid, malefic acid,
methanesulfonic acid,
succinic acid and citric acid. Basic addition salts can be prepared in situ
during the final
isolation and purification of the compounds of formula (l~, or separately by
reacting
carboxylic acid moieties with a suitable base such as the hydroxide, carbonate
or
bicarbonate of a pharmaceutically acceptable metal cation or with ammonia, or
an
organic primary, secondary or tertiary amine. Pharmaceutically acceptable
salts include,
but are not limited to, canons based on the alkali and alkaline earth metals,
such as
sodium, lithium, potassium, calcium, magnesium, aluminum salts and the like,
as well as
nontoxic ammonium, quaternary ammonium, and amine cations, including, but not
limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine,
dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. Other
representative organic amines useful for the formation of base addition salts
include
diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, and
the like.
The term "pharmaceutically acceptable prodrugs" as used herein refers to those
prodrugs of the compounds of the present invention which are, within the scope
of sound
medical judgment, suitable for use in contact with the tissues of humans and
lower
animals without undue toxicity, irntation, allergic response, and the like,
commensurate
with a reasonable benefit/risk ratio, and effective fox their intended use, as
well as the
zwitterionic forms, where possible, of the compounds of the invention. The
term
"prodrug" refers to compounds that are rapidly transformed in vivo to yield
the parent
-24-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
compound of the above formula, fox example by hydrolysis in blood. A thorough
discussion is provided in Higuchi, T., and V. Stella, "Pro-drugs as Novel
Delivery
Systems," A.C.S. Symposium Series 14, and in "Bioreversible Carriers in Drug
Design,"
in Edward B. Roche (ed.), American Pharmaceutical Association, Pergamon Press,
1987,
both of which are incorporated herein by reference.
The term "cancer" refers to cancer diseases that cam be beneficially treated
by the
inhibition of KSP, including, for example, solid cancers, such as carcinomas
(e.g., of the
lungs, pancreas, thyroid, bladder or colon); myeloid disorders (e.g., myeloid
leukemia);
and adenomas (e.g., villous colon adenoma). Cancer is a proliferative disease.
The compounds of the invention are useful in vitro or in vivo in inhibiting
the
growth of cancer cells. The compounds may be used alone or in compositions
together
with a pharmaceutically acceptable carrier or excipient. Suitable
pharmaceutically
acceptable carriers or excipients include, for example, processing agents and
drug
delivery modifiers and enhancers, such as, for example, calcium phosphate,
magnesium
stearate, talc, monosaccharides, disaccharides, starch, gelatin, cellulose,
methyl cellulose,
sodium carboxymethyl cellulose, dextrose, hydroxypropyl-[3-cyclodextrin,
polyvinyl-
pyrrolidinone, low melting waxes, ion exchange resins, and the like, as well
as
combinations of any two or more thereof. Other suitable pharmaceutically
acceptable
excipients are described in "Remington's Pharmaceutical Sciences," Mack Pub.
Co., New
Jersey, 1991, incorporated herein by reference.
Effective amounts of the compounds of the invention generally include any
amount sufficient to detectably inhibit KSP activity by any of the assays
described herein,
by other KSP activity assays known to those having ordinary skill in the axt,
or by
detecting an inhibition or alleviation of symptoms of cancer.
The amount of active ingredient that may be combined with the carrier
materials
to produce a single dosage form will vary depending upon the host treated and
the
particular mode of administration. It will be understood, however, that the
specific dose
level for any particular patient will depend upon a vaxiety of factors
including the activity
of the specific compound employed, the age, body weight, general health, sex,
diet, time
of administration, route of administration, rate of excretion, drug
combination, and the
severity of the particular disease undergoing therapy. The therapeutically
effective
amount for a given situation can be readily determined by routine
experimentation and is
within the skill and judgment of the ordinary clinician.
-25-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
For purposes of the present invention, a therapeutically effective dose will
generally be a total daily dose administered to a host in single or divided
doses may be in
amounts, for example, of from 0.001 to 1000 mglkg body weight daily and more
preferred from 1.0 to 30 mglkg body weight daily. Dosage unit compositions may
contain such amounts of submultiples thereof to make up the daily dose.
The compounds of the present invention may be administered orally,
parenterally,
sublingually, by aerosolization or inhalation spray, rectally, or topically in
dosage unit
formulations containing conventional nontoxic pharmaceutically acceptable
carriers,
adjuvants, and vehicles as desired. Topical administration may also involve
the use of
transdermal administration such as transdennal patches or ionophoresis
devices. The
term parenteral as used herein includes subcutaneous injections, intravenous,
intramuscular, intrasternal injection, or infusion techniques.
Injectable preparations, for example, sterile injectable aqueous or oleaginous
suspensions may be formulated according to the known art using suitable
dispersing or
wetting agents and suspending agents. The sterile injectable preparation may
also be a
sterile injectable solution or suspension in a nontoxic parenterally
acceptable diluent or
solvent, for example, as a solution in 1,3-propanediol. Among the acceptable
vehicles
and solvents that may be employed are water, Ringer's solution, and isotonic
sodium
chloride solution. In addition, sterile, fixed oils are conventionally
employed as a solvent
or suspending medium. For this purpose any bland fixed oil may be employed
including
synthetic mono- or di-glycerides. In addition, fatty acids such as oleic acid
find use in the
preparation of injectables.
Suppositories for rectal administration of the drug can be prepared by mixing
the
drug with a suitable nonirritating excipient such as cocoa butter and
polyethylene glycols,
which are solid at ordinary temperatures but liquid at the rectal temperature
and will
therefore melt in the rectum and release the drug.
Solid dosage forms for oral administration may include capsules, tablets,
pills,
powders, and granules. In such solid dosage forms, the active compound may be
admixed with at least one inert diluent such as sucrose lactose or starch.
Such dosage
forms may also comprise, as is normal practice, additional substances other
than inert
diluents, e.g., lubricating agents such as magnesium stearate. In the case of
capsules,
tablets, and pills, the dosage forms may also comprise buffering agents.
Tablets and pills
can additionally be prepared with enteric coatings.
-26-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Liquid dosage forms for oral administration may include pharmaceutically
acceptable emulsions, solutions, suspensions, syrups, and elixirs containing
inert diluents
commonly used in the art, such as water. Such compositions may also comprise
adjuvants, such as wetting agents, emulsifying and suspending agents,
cyclodextrins, and
sweetening, flavoring, and perfuming agents.
The compounds of the present invention can also be administered in the form of
liposomes. As is known in the art, liposomes are generally derived from
phospholipids or
other lipid substances. Liposomes are formed by mono- or multi-lamellar
hydrated liquid
crystals that are dispersed in an aqueous medium. Any non-toxic,
physiologically
acceptable and metabolizable lipid capable of forming liposomes can be used.
The
present compositions in liposome form can contain, in addition to a compound
of the
present invention, stabilizers, preservatives, excipients, and the like. The
preferred lipids
are the phospholipids and phosphatidyl cholines (lecithins), both natural and.
synthetic.
Methods to form liposomes are known in the art. See, for example, Prescott
(ed.),
"Methods in Cell Biology," Volume XIV, Academic Press, New York, 1976, p. 33
et seq.
While the compounds of the invention can be administered as the sole active
pharmaceutical agent, they can also be used in combination with one or more
other agents
used in the treatment of cancer. Representative agents useful in combination
with the
compounds of the invention for the treatment of cancer include, for example,
irinotecan,
topotecan, gemcitabine, gleevec, herceptin, 5-fluorouracil, leucovorin,
carboplatin,
cisplatin, taxanes, tezacitabine, cyclophosphamide, vinca alkaloids, imatinib,
anthracyclines, rituximab, trastuzumab, topoisomerase I inhibitors, as well as
other cancer
chemotherapeutic agents.
The above compounds to be employed in combination with the compounds of the
invention will be used in therapeutic amounts as indicated in the Physicians'
Desk
Reference (PDR) 47th Edition (1993), which is incorporated herein by
reference, or such
therapeutically useful amounts as would be known to one of ordinary skill in
the art.
The compounds of the invention and the other anticancer agents can be
administered at the recommended maximum clinical dosage or at lower doses.
Dosage
levels of the active compounds in the compositions of the invention may be
varied so as
to obtain a desired therapeutic response depending on the route of
administration, severity
of the disease and the response of the patient. The combination can be
administered as
separate compositions or as a single dosage form containing both agents. When
-27-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
administered as a combination, the therapeutic agents can be formulated as
separate
compositions, which are given at the same time or different times, or the
therapeutic
agents, can be given as a single composition.
Antiestrogens, such as tamoxifen, inhibit breast cancer growth through
induction
of cell cycle arrest, that requires the action of the cell cycle inhibitor
p27Kip. Recently, it
has been shown that activation of the Ras-Raf MAP Kinase pathway alters the
phosphorylation status of p27Kip such that its inhibitory activity in
arresting the cell
cycle is attenuated, thereby contributing to antiestrogen resistance (Donovan,
et al,
J. Biol. C7zem. 276:40888, 2001). As reported by Donovan et al., inhibition of
MAPK
signaling through treatment with MEK inhibitor changed the phosphorylation
status of
p27 in hormone refactory breast cancer cell lines and in so doing restored
hormone
sensitivity. Accordingly, in one aspect, the compounds of formula (~ may be
used in the
treatment of hormone dependent cancers, such as breast and prostate cancers,
to reverse
hormone resistance commonly seen in these cancers with conventional anticancer
agents.
In hematological cancers, such as chronic myelogenous leukemia (CML),
chromosomal translocation is responsible for the constitutively activated BCR-
AB 1
tyrosine kinase. The afflicted patients are responsive to gleevec, a small
molecule
tyrosine kinase inhibitor, as a result of inhibition of Ab 1 kinase activity.
However, many
patients with advanced stage disease respond to gleevec initially, but then
relapse later
due to resistance-conferring mutations in the Abl kinase domain. Ira vitro
studies have
demonstrated that BCR-Av1 employs the Raf kinase pathway to elicit its
effects. In
addition, inhibiting more than one kinase in the same pathway provides
additional
protection against resistance-conferring mutations. Accordingly, in another
aspect of the
invention, the compounds of formula (I) are used in combination with at least
one
additional agent, such as gleevec, in the treatment of hematological cancers,
such as
chronic myelogenous leukemia (CML), to reverse or prevent resistance to the at
least one
additional agent.
In another aspect of the invention, kits that include one or more compounds of
the
invention are provided. Representative kits include a compound of formula (I)
and a
package insert or other labeling including directions for treating a cellular
proliferative
disease by administering an KSP inhibitory amount of the compound.
_28_



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
The present invention will be understood more readily by reference to the
following examples, which are provided by way of illustration and are not
intended to be
limiting of the present invention.
EXAMPLES
Referring to the examples that follow, compounds of the present invention were
synthesized using the methods described herein, or other methods, which are
well known
in the art.
The compounds and/or intermediates were characterized by high performance
liquid chromatography (HPLC) using a Waters Millenium chromatography system
with a
2690 Separation Module (Milford, MA). The analytical columns were Alltima C-18
reversed phase, 4.6 x 250 mm from Alltech (Deerfield, IL). A gradient elution
was used,
typically starting with 5% acetonitrile/95% water and progressing to 100%
acetonitrile
over a period of 40 minutes. All solvents contained 0.1% trifluoroacetic acid
(TFA).
Compounds were detected by ultraviolet light (UV) absorption at either 220 or
254 nm.
HPLC solvents were from Burdick and Jackson (Muskegan, MI), or Fisher
Scientific
(Pittsburgh, PA). In some instances, purity was assessed by thin layer
chromatography
(TLC) using glass or plastic backed silica gel plates, such as, for example,
Baker-Flex
Silica Gel 1B2-F flexible sheets. TLC results were readily detected visually
under
' ultraviolet light, or by employing well known iodine vapor and other various
staining
techniques.
Mass spectrometric analysis was performed on one of two LCMS instruments: a
Waters System (Alliance HT HPLC and a Micromass ZQ mass spectrometer; Column:
Eclipse XDB-C18, 2.1 x 50 mm; solvent system: 5-95% (or 35-95%, or 65-95% or
95-95%) acetonitrile in water with 0.05%TFA; flow rate 0.8 mL/min; molecular
weight
range 500-1500; cone Voltage 20 V; column temperature 40 °C) or a
Hewlett Packard
System (Series 1100 HPLC; Column: Eclipse XDB-C18; 2.1 x 50 mm; solvent
system:
1-95% acetonitrile in water with 0.05%TFA; flow rate 0.4 mL/min; molecular
weight
range 150-850; cone Voltage 50 V; column temperature 30 °C). All masses
were reported
as those of the protonated parent ions.
GCMS analysis is performed on a Hewlett Packard instrument (HP6890 Series
gas chromatograph with a Mass Selective Detector 5973; injector volume: 1 p,L;
initial
column temperature: 50 °C; final column temperature: 250 ° tramp
time: 20 minutes;
-29-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
gas flow rate: 1 mL/min; column: 5% phenyl methyl siloxane, Model
No. HP 190915-443, dimensions: 30.0 m x 25 m x 0.25 m).
Nuclear magnetic resonance (NMR) analysis was performed on some of the
compounds with a Varian 300 MHz NMR (Palo Alto, CA). The spectral reference
was
either TMS or the known chemical shift of the solvent. Some compound samples
were
run at elevated temperatures (e.g., 75°C) to promote increased sample
solubility.
The purity of some of the invention compounds is assessed by elemental
analysis
(Desert Analytics, Tucson, AZ)
Melting points are determined on a Laboratory Devices Mel-Temp apparatus
(Holliston, MA).
Preparative separations were carried out using a Flash 40 chromatography
system
and KP-Sil, 60A (Biotage, Charlottesville, VA), or by flash column
chromatography
using silica gel (230-400 mesh) packing material, or by HPLC using a C-18
reversed
phase column. Typical solvents employed for the Flash 40 Biotage system and
flash
column chromatography were dichloromethane, methanol, ethyl acetate, hexane,
acetone,
aqueous hydroxyamine, and triethyl amine. Typical solvents employed for the
reverse
phase HPLC were varying concentrations of acetonitrile and water with
0.1% trifluoroacetic acid.
The following are abbreviations used in the examples:
AcOH: Acetic acid
aq: Aqueous
ATP: Adenosine triphosphate
9-BBN 9-Borabicyclo[3.3.1]nonane
Boc: tert-butoxycarbonyl
Celite Filter agent
DAP or Dap: Diaminopropionate
DCM: Dichloromethane
DEAD: Diethyl azodicarboxylate
DIEA: Diisopropylethylamine


DMAP 4-Dimethylaminopyridine


DME: 1,2-Dimethoxyethane


DMF: N,N-Dimethylformamide


DMSO: Dimethyl sulfoxide


-30-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
DPPA: biphenyl phosphoryl azide


Et3N: Triethylamine


EDC: N-(3-Dimethylaminopropyl)-N'-ethylcarbodiimide


EDCI: 1-(3-Dimethylaminopropyl)3-ethylcarbodiimide


EtOAc: Ethyl acetate


EtOH: Ethanol


Fmoc: 9-Fluorenylmethoxycarbonyl


Gly-OH: Glycine


HATU: O-(7-Azabenzotriaazol-1-yl)-N,N,N'N'-tetramethyluronium


hexafluorophosphate


HBTU: 2-(1H-Benzotriazol-1-yl)-1,1,3,3-tetramethyluronium


hexafluorophosphate


Hex: Hexane


HOBt: Butyl alcohol


HOBT: 1-Hydroxybenzotriazole


HPLC: High pressure liquid chromatography


NIS N-Iodosuccinimide


ICSO value: The concentration of an inhibitor that causes
a 50% reduction


in a measured activity.


iPrOH: Isopropanol


LC/MS: Liquid chromatographylmass spectrometry


LRMS: LoW resolution mass spectrometry


MeOH: Methanol


NaOMe: Sodium methoxide


nm: Nanometer


NMP: N-Methylpyrrolidone


PPA Polyphosphoric acid


PPh3: Triphenyl phosphine


PTFE Polytetrafluoroethylene


RP-HPLC: Reversed-phase high-pressure liquid chromatography


RT: Room temperature


sat: Saturated


TEA: Triethylamine


-31-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
TFA: Trifluoroacetic acid


THF: Tetrahydrofuran


Thr: Threonine


TLC: Thin layer chromatography


Trt-Br: Tert-butyl bromide


Nomenclature for the Example compounds was provided using ACD Name
version 5.07 software (November 14, 2001) available from Advanced Chemistry
Development, Inc. Some of the compounds and starting materials were named
using
standard ICTPAC nomenclature.
It should be understood that the organic compounds according to the invention
may exhibit the phenomenon of tautomerism. As the chemical structures within
this
specification can only represent one of the possible tautomeric forms, it
should be
understood that the invention encompasses any tautomeric form of the drawn
structure.
It is understood that the invention is not limited to the embodiments set
forth
herein for illustration, but embraces all such forms thereof as come within
the scope of
the above disclosure.
Example 1
Synthesis of N (3-aminopropyl)-N f 1-(3-benzyl-7-chloro-4-oxo-3,4-
dihydroguinazolin 2 yl)-2-(dimethylamino)-2-oxoethyl~-4-methylbenzamide
Step 1: 3-benzyl-7-chloro-2-methylquinazolin-4 (3I~-one.
POCI3
microwave
C02H H ~ I 150°C, 10min. O
+ O N
N
CI NH2
CI N CH3
2-Amino-4-chlorobenzoic acid (250 mg, 1 eq.) and N-benzylacetamide (261 mg,
1.2 eq.) were dissolved in phosphorus oxychloride (2 ml). The reaction was
placed in the
microwave for 10 minutes at 150°C. The reaction was quenched by pouring
the mixture
into ice. The crude product was extracted into ethyl acetate. The ethyl
acetate layer was
washed with water, 10% aqueous sodium hydroxide, dried with magnesium sulfate,
filtered, and concentrated to yield 3-benzyl-7-chloro-2-methylquinazolin-4
(3I~-one as
an orange solid.
-32-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Step 2: 2-(3-benzyl-7-chloro-4-oxo-3, 4-dihydroquinazolin-2-yl)-N,N dimethyl-
acetamide.
I\ I\
O / O LHMDS O
THF
CI~N~ ~$°C \ I N O
CI N CH3 I CI N~N~
I
3-Benzyl-7-chloro-2-methylquinazolin-4 (3I~-one (2 g, 1 eq.) was dissolved in
tetrahydrofuran (20 ml). The solution was cooled to -78°C for 15
minutes. Lithium
hexamethyl disilazide (LHMDS) was added slowly and the mixture was stirred at -
78°C
for 45 minutes. Dimethylcarbamyl chloride was added and the mixture was
stirred at
-78°C for 1 h. The reaction was allowed to warm to room temperature and
stirred for 3 h.
The solvent was evaporated and the resulting solid was dissolved in ethyl
acetate. The
ethyl acetate layer was washed with water, saturated sodium bicarbonate,
saturated
sodium chloride, dried over magnesium sulfate, filtered and concentrated. The
crude
product was purified by flash chromatography to give 2-(3-benzyl-7-chloro-4-
oxo-3,
4-dihydroquinazolin-2-yl)-N,N dimethylacetamide (1.2 g) as a solid.
Step 3: 2-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-bromo-N,N
dimethylacetamide.
O I ~ Br2 O
NaOAc
N O AcOH / ~N O
I ~ I
CI \ N~N~ CI ~ N~N~
Br I
To a mixture 2-(3-benzyl-7-chloro-4-oxo-3, 4-dihydroquinazolin-2-yl)-N,N
dimethylacetamide (1.1 g, 1 eq.) and sodium acetate (761 mg, 3 eq.) in acetic
acid
(15 ml), . was added bromine (158 ul, 1 eq.) and the reaction was stirred at
room
temperature for 4 h. Water was added and the product extracted into ethyl
acetate. The
ethyl acetate layer was washed several times with water, saturated sodium
bicarbonate,
saturated sodium chloride, dried over magnesium sulfate, filtered, and
concentrated to
-33-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
give 2-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-bromo-N,N
dimethylacetamide (1.44 g) as a yellow solid.
Step 4: tent-butyl 3-{[1-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
2-
(dimethylamino)-2-oxoethyl] amino}propylcarbamate.
O
o ~ ~ ~ N o
H N~NHBoc
N O 2 CI \ N~N~
CI ~ N~N~ HN
Br
NHBoc
To a solution of 2-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-
bromo-N,N dimethylacetamide (1.4 g, 1 eq.) in of N,N-dimethylformamide (10
ml), was
added N-Boc-1,3-diaminopropane (2.~ g, 5 eq.). The reaction was stirred at
room
temperature for 4 h and then heated to 40°C. The mixture was stirred at
40°C overnight.
Water and ethyl acetate were added to the mixture. The organic layer was
washed with
saturated sodium bicarbonate, saturated sodium chloride, dried over magnesium
sulfate,
filtered, and concentrated. The crude product was purified by flash
chromatography to
give test-butyl 3-{[1-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-
(dimethylamino)-2-oxoethyl]amino)propylcarbamate (1 g) as a pale yellow solid.
Step 5: teat-butyl 3-[[1-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
2-
(dimethylamino)-2-oxoethyl](4-methylbenzoyl)amino]propylcarbamate.
i
0
0
COCI pMF ~ I ~N O
N O I ~ ~ CI \ N~N~
CI ~ N~N~ + / ~' O N
HN
CH3
NHBoc
NHBoc
CH3
-34-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
To a solution of tent-butyl 3-{[1-(3-benzyl-7-chloro-4-oxo-3,4-
dihydroquinazolin-
2-yl)-2-(dimethylamino)-2-oxoethyl]amino}propylcarbamate (500 mg, 1 eq.) in
methylene chloride (5 ml), was added 4-methyl benzoyl chloride (380 ul, 3 eq.)
and
triethylamine (660 ul, 5 eq.). The reaction mixture was stirred at room
temperature
overnight. The solvent Was evaporated and the crude product was dissolved in
ethyl
acetate. Ethyl acetate layer was washed with water, saturated sodium
carbonate,
saturated sodium chloride, dried over magnesium sulfate, filtered, and
evaporated to yield
crude tent-butyl 3-[[1-(3-benzyl-7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-
(dimethylamino)-2-oxoethyl](4-methylbenzoyl)amino]propylcarbamate.
Step 6: N (3-aminopropyl)-N [1-(3-benzyl-7-chloro-4-oxo-3,4-dihydro-
quinazolin-2-yl)-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide.
w
/ o /
/ N O ~~% trifluoroacetic acid / ~N O
in CH2CI2
CI '~ N N~ ~ CI N~N
O N
O N
NHBoc w ' NH2
CH3 CH3
To a solution of tent-butyl 3-[[1-(3-benzyl-7-chloro-4-oxo-3,4-
dihydroquinazolin-
2-yl)-2-(dimethylamino)-2-oxoethyl](4-methylbenzoyl)amino]propylcarbamate (500
mg),
was added 20% solution of trifluoroacetic acid in methylene chloride at room
temperature. The mixture was stirred at room temperature overnight. The
solvent was
removed under reduced pressure. The crude product was purified by flash
chromatography to give N (3-aminopropyl)-N [1-(3-benzyl-7-chloro-4-oxo-3,4-
dihydroquinazolin-2-yl)-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide (336
mg) as
a white solid.
-35-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Example 2
Synthesis of N (3-aminopropyl)-N f 1-[7-chloro-3-(3-methylbenzyl)-4-oxo-3,4-
dih,~quinazolin-2-~]-2-(dimethyl~ino)-2-oxoethyll-4-methylbenzamide
Step 1: 2-(7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-N,N dimethylacetamide.
O
C02CH3 O
HCI in dioxane ~ ~NH O
+ NC~N~ rt, overnight I / ~ i
CI NH2 I CI N N
To a mixture of 2-amino-4-chloro methyl benzoate (5 g, 1 eq.) and N,N-
dimethylamino acetamide (3.63 g, 1.2 eq.), was added a solution of 4M HCl in
dioxane
(40 ml). The reaction was stirred at room temperature overnight. Dioxane was
removed at
reduced pressure and the resulting solid residue was triturated with cold
water. The
aqueous solution was basified with 10% ammonium hydroxide. The precipitate was
collected, washed with more cold water, washed with ether and dried to yield
2-(7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-N,N dimethylacetamide 7.4 g (96
%) as a
white solid.
Step 2: 2-bromo-2-(7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-N,N dimethyl-
acetamide.
O
O
~NH O
NH O NaOAc
~ ~ ~ + Br2 c CI f ~ N~N~
CI N~N rt, overnight Br I
I
To a mixture of 2-(7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-N,N
dimethylacetamide and sodium acetate (930 mg, 3 eq.) in acetic acid (15 ml),
was added
bromine (190 ul, 1 eq.). The reaction mixture was stirred at room temperature
overnight.
Water was added and the product was extracted into ethyl acetate. The ethyl
acetate layer
was washed several times with water, saturated sodium bicarbonate, saturated
sodium
chloride, dried over magnesium sulfate, filtered, and concentrated to give 2-
bromo-2-(7-
chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-N,N dimethylacetamide (0.95 g) as a
brown
solid.
-36-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Step 3: tent-butyl 3-~[1-(7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-
(dimethylamino)-2-oxoethyl]amino)propylcarbamate.
O
O ' ~ ~NH O
CI / N~N~
~NH O
+ H2N~NHBoc gp C, 0.5h HN
CI /' N~N
Br
NHBoc
To a solution of 2-bromo-2-(7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-N,N
dimethylacetamide (950 mg, 1 eq.) in N,N-dimethylformamide (10 ml), was added
N-Boc-1,3-diaminopropane (2.4 g, 5 eq.). The reaction was stirred at 60
°C for
30 minutes. Water and ethyl acetate was added. Ethyl acetatelayer was washed
with
saturated sodium bicarbonate, saturated sodium chloride, dried over magnesium
sulfate,
filtered, and concentrated to give tent-butyl 3- f [1-(7-chloro-4-oxo-3,4-
dihydroquinazolin-
2-yl)-2-(dimethylamino)-2-oxoethyl]amino~propylcarbamate (1.2 g) as a pale
orange
solid.
Step 4: 2-[1-[ f 3-[(tef~t-butoxycarbonyl)amino]propyl~(4-methylbenzoyl)amino]-

2-(dimethylamino)-2-oxoethyl]-7-chloro-3,4-dihydroquinazolin-4-yl4-
methylbenzoate.
CH3
O
COCI O O
~NH O ~ NH O
/ ~ ~ + ~ TEA
CI N N ~ / CH2C12 CI I / N N~
HN ~ rt, overnight
CH3 O N
NHBoc /
NHBoc
CH3
To a solution of tent-butyl 3-{[1-(7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
2-
(dimethylamino)-2-oxoethyl]amino)propylcarbamate (1.2 g, 1 eq.) methylene
chloride
-37-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
920 ml) at 0 °C, was added 4-methyl benzoyl chloride (1.1 ml, 3 eq.)
and triethylamine
(1.9 ml, 5 eq.). The reaction was allowed to warm up to room temperature and
stirred at
room temperature overnight. The solvent was evaporated and the crude product
was
dissolved in ethyl acetate. Ethyl acetate layer was washed with water,
saturated sodium
carbonate, saturated sodium chloride, dried over magnesium sulfate, filtered,
and
evaporated to yield 2-[1-[{3-[(tent-butoxycarbonyl)amino]propyl~(4-
methylbenzoyl)amino]-2-(dimethylamino)-2-oxoethyl]-7-chloro-3,4-
dihydroquinazolin-
4-yl 4-methylbenzoate (2.3 g) as a brown oil.
Step 5: tent-butyl 3-[[1-(7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-
(dimethyl-
amino)-2-oxoethyl](4-methylbenzoyl)amino]propylcarbamate.
CH3
O
'NH O
O O CI ~ N~N~
NH O LiOH O N
CI I ~ N Ni THF / H2O
rt, 3h
O N
NHBoc
CH3
NHBoc
CH3
To a solution of 2-[1-[~3-[(tart-butoxycarbonyl)amino]propyl}(4-
methylbenzoyl)amino]-2-(dimethylamino)-2-oxoethyl]-7-chloro-3,4-
dihydroquinazolin-
4-yl 4-methylbenzoate in a 1:1 mixture of tetrahydrofuran and water (20 ml),
was added
lithium hydroxide (284 mg, 2 eq.) at room temperature. The mixture was stirred
at room
temperature for 3 h. The reaction mixture was acidified with 1M of aqueous
hydrogen
chloride to pH 6 - 7 and later extracted into methylene chloride. The organic
layer was
washed with more water, dried over magnesium sulfate, filtered and
concentrated. The
product was purified by flash chromatography to give tart-butyl 3-[[1-(7-
chloro-4-oxo-
3,4-dihydroquinazolin-2-yl)-2-(dimethylamino)-2-oxoethyl](4-methylbenzoyl)-
amino]-
propylcarbamate (257 mg) as a yellow solid.
-3 8-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Step 6: te~~t-butyl 3-[[1-[7-chloro-3-(3-methylbenzyl)-4-oxo-3,4-dihydro-
quinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl](4-methylbenzoyl)amino]propyl-
carbamate.
CH3
O
O
NH O ~ ~Br
/ ~ / + ~ / K2C03 ~ ~ N O
CI N~N '
CH ~~ overnight CI / N~N~
O N s O N
NHBoc
W ~ NHBoc
CH3 CH3
To a solution of tent-butyl 3-[[1-(7-chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-
2,-
(dimethylamino)-2-oxoethyl](4-methylbenzoyl)amino]propylcarbamate (29 mg, 1
eq.) in
dimethyl formamide (1 ml), was added 3-methylbenzyl bromide (8 p,l, 1.2 eq.)
and
potassium carbonate (22 mg, 3 eq.). The reaction mixture was stirred at room
temperature
overnight. Water was added and the material was extracted into ethyl acetate.
Ethyl
acetate layer was washed with water, saturated sodium carbonate, saturated
sodium
chloride, dried over magnesium sulfate, filtered, and evaporated. The product
was
purified by flash chromatography to give tef~t-butyl 3-[[1-[7-chloro-3-(3-
methylbenzyl)-4-
oxo-3,4-dihydroquinazolin-2-yl]-2.-(dimethylamino)-2-oxoethyl](4-
methylbenzoyl)amino]propylcarbamate.
-3 9-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Step 7: N (3-aminopropyl)-N [1-[7-chloro-3-(3-methylbenzyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide.
~ CH3 I ~ CH3
O / O
~N O ~ ~N O
CI I / N~N~ CH2CI2 CI I / N~N~
O N ~ rt, 2h O N
NHBoc ~ ~ NH2
CH3 CH3
To a solution of test-butyl 3-[[1-[7-chloro-3-(3-methylbenzyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl](4-
methylbenzoyl)amino]propyl-
carbaanate, was added 20% solution of trifluoroacetic acid in methylene
chloride at room
temperature. The reaction mixture was stirred at room temperature for 2 h. The
solvent
was removed under reduced pressure. The crude product was purified by reverse
phase
HPLC to give N (3-aminopropyl)-N [1-[7-chloro-3-(3-methylbenzyl)-4-oxo-3,4-
dihydroquinazolin-2-yl]-2-(dimethylamino)-2-oxoethyl]-4-methylbenzamide (9 mg)
as
TFA salt.
-40-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Example 3
Representative Q_uinazolinone Compounds
Representative quinazolinone compound compounds of the invention are shown
in Table 1. In Table 1, MH+ refers to the molecular ion observed by mass
spectrometry.
Table 1. Representative Quinazolinone Compounds.
Com ound Structure MH+ Name
1 I ' 458.0 ethyl (3-benzyl-7-chloro-4-oxo-3,4-
o ~ dihydroquinazolin-2-yl){[3-
(dimethylamino)propyl]amino}acetate
ci ' ni~o~°H,
NH
H,°. N. °H,
641.0 ethyl (3-benzyl-7-chloro-4-oxo-3,4-
dihydroquinazolin-2-yl){(4-
bromobenzoyl)[3-
CI ' N O~CH,
~N o (dimethylamino)propyl]amino}acetate
'i
H,C.N.CH,
Br
3 i ' 669.0 ethyl 2-(3-benzyl-7-chloro-4-oxo-3,4-
° ' dihydroquinazolin-~-yl)-2-{(4-
N ~Ha bromobenzoyl)[3-
a ~-'~ (dimethylamino)propyl]amino}butanoate
H,C-N _ CH3
CH ~
Br
4 i ~ 576.1 ethyl (3-benzyl-7-chloro-4-oxo-3,4-
dihydroquinazolin-2-yl)[[3-
ol ~ ' N °~rH, (dimethylamino)propyl](4-
O N~N.CH, methylbenzoyl)amino]acetate
'i
'
cH,
5 I ' 641.0 ethyl (3-benzyl-7-chloro-4-oxo-3,4-
o ~ dihydroquinazolin-2-yl){(3-
N ° bromobenzoyl)[3-
°I ' ri~o~cH, (dimethyfamino)propyl]amino} acetate
0 N~'N.°Hs
i
Br' v
-41-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Com ound Structure MN+ Name
g I ' 641.0 ethyl (3-benzyl-7-chloro-4-oxo-3,4-
o ~ dihydroquinazolin-2-yl)((2-
~ ' N ° bromobenzoyl)[3-
cl ~ N~O~CH3 (dimethylamino)propyl]amino}acetate
O N~N.C~
Br~
7 ~ ~ 548.1 ethyl [(3-aminopropyl)(4-
° ' methylbenzoyl)amino](3-benzyl-7-chloro
-oxo-3,4-dihydroquinazolin-2-yl)acetate
CI ~ N O~CH3
0 N~NHi
'
CH,
g I ' 576.1 ethyl (3-benzyl-7-chloro-4-oxo-3,4-
o dihydroquinazolin-2-yl)[[3-
~ ' N ° (dimethylamino)propyl](3-
' J~l. ~
CI O N~IJH°H' methylbenzoyl)amino]acetate
~'I
H C"V
3
g I ' 576.1 ethyl (3-benzyl-7-chloro-4-oxo-3,4-
o dihydroquinazolin-2-yl)[[3-
~ ' N ° (dimethylamino)propyl](2-
' J~l ~
CI N O CH3 methylbenzoyl)amino]acetate
O N~N.CH3
~C
~ ~ 640.0 N-[1-(3-benzyl-7-chloro-4-oxo-3,4-
' dihydroquinazolin-2-yl)-2-(ethylamino)-2-
N o oxoethyl]-4-bromo-N-[3-
ol ' ~p~cH~ (dimethyfamino)propyl]benzamide
~H,C,N,CH3
6r
11 ~ ~ 655.0 isopropyl (3-benzyl-7-chloro-4-oxo-3,4-
dihydroquinazolin-2-yl)((4-
i ' ~ ~' bromobenzoyl)[3-
CI ~ NO N O CH3 (dimethylamino)propyl]amino}acetate
~H'C.N,CH3
er
-42-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Com oundStructure MH+ Name


12 i ~ 590.1sopropyl (3-benzyl-7-chloro-4-oxo-3,4-
i


' dihydroquinazolin-2-yl)[[3-
~ \ l
(4
i
th
l
i


N~o~H, ( ]
m -
am
no)propy
y
me
d


N methylbenzoyl)amino]acetate


'i
~H3C.N,CH'
CHI


13 i ~ 626.9sopropyl [(3-aminopropyl)(4-
i


' bromobenzoyl)amino](3-benzyl-7-chloro-4-
~ li
cet
te
i
2-
l
4
dih
d
3


' N~O~CH roqu
cl nazo
3 n-
y
)a
a
-
y
oxo-
,


O N'
l'l'
~ NHa


Br


14 i ~ 562.1sopropyl [(3-aminopropyl)(4-
i


' methylbenzoyl)amino](3-benzyl-7-chloro-
I t
2
l
t
i
li
dih
d


' N~O~CH -y
of )ace
3 a
e
roqu
nazo
n-
y
-oxo-3,4-


O N
'
~ NHi



CH,


15 ~ ~ 640.0N-(3-aminopropyl)-N-[1-(3-benzyl-7-


' chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-
(diethylamino)-2-oxoethyl]-4-


CI ' N NCH,


O N~LCH3 bromobenzamide


NHi


Br


1g i ~ 575.1N-(3-aminopropyl)-N-[1-(3-benzyl-7-
'


chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-
I (diethylamino)-2-oxoethyl]-4-


' N~N~CH
of
J


O N' LCH3 methylbenzamide
l'


l'
I NHz


CH,


17 i ~ 638.0N-(3-aminopropyl)-N-[1-(3-benzyl-7-
'


chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-
CI N N oxo-2-pyrrolidin-1-ylethyl]-4-


N V bromobenzamide
'
~ ~ NH=



Br


-43-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Com oundStructure MH+ Name


18 i ~ 593.5N-(3-aminopropyl)-N-j1-(3-benzyl-7-
~ chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-
I i ~ N~N oxo-2-pyrrolidin-1-ylethyl]-4-


~ chlorobenzamide
N
I NHz


CI


19 i ~ 654.0N-(3_aminopropyl)-N-[1-(3-benzyl-7-
~ chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-
CI I ' N~N~ morpholin-4-yl-2-oxoethyi]-4-
o N~~o bromobenzamide
~ NHz


Br


2p i ~ 609.5N-(3-aminopropyl)-N-[1-(3-benzyl-7-
chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-
I ' ~~~~ morpholin-4-yl-2-oxoethyl]-4-
chlorobenzamide
I ~ Hz


CI


21 i ~ 611.9N-(3-aminopropyl)-N-[1-(3-benzyl-7-
chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-
CI I ' N N N-CN' (dimethylamino)-2-oxoethyi]-4-
bromobenzamide


~ ~ Hz



22 ~ ~ 567.5N-(3-aminopropyl)-N-[1-(3-benzyl-7-
~ chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-
, I ' N~N~CH' (dimethylamino)-2-oxoethyl]-4-
O N~CH3 chlorobenzamide


s
W ~ NHz


CI


23 i ~ 573.1N-(3-aminopropyl)-N-[1-(3-benzyl-7-
chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-
I ~ ' N~N~ oxo-2-pyrrolidin-1-ylethyl]-4-


N methylbenzamide
~ ~ NHz


CH,


-44-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Com oundStructure MH-~ Name


24 i a 547.1N-(3-aminopropyl)-N-[1-(3-benzyl-7-
chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-
~I ~ ~ N N O NCH, (dimethylamino)-2-oxoethyl]-4-
~~H~ methylbenzamide


~ ~ NHi


CH,


25 i ~ 626.0N-(3-aminopropyl)-N-[1-[3-(2-
' B~ bromobenzyl)-7-chloro-4-oxo-3,4-
~N O
ci ~ ' N~N'H' dihydroquinazolin-2-yl]-2-(dimethylamino)-
o 'N~~H, 2-oxoethyl]-4-methylbenzamide


w ~ NH,


CH,


2g i ~ 581.5N-(3-aminopropyl)-N-[1-[7-chloro-3-(2-
~I chlorobenzyl)-4-oxo-3,4-
~N O
~ ~ ' N~N'H' dihydroquinazolin-2-yl]-2-(dimethylamino)-
O NCH, 2-oxoethyl]-4-methylbenzamide


w ~ NHz


CH,


27 ~ ~~ 581.5N-(3-aminopropyl)-N-[1-[7-chloro-3-(3-


~ chlorobenzyl)-4-oxo-3,4-
~N O
~ ~ ' N~N'CH' dihydroquinazolin-2-yl]-2-(dimethylamino)-
O NCH, 2-oxoethyl]-4-methylbenzamide


~ ~ NHz


CH,


28 ~ F 581.5N-(3-aminopropyl)-N-[1-[7-chloro-3-(3-


luorobenzyl)-4-oxo-3,4-dihydroquinazolin-
N 2-yl]-2-(dimethylamino)-2-oxoethyl]-4-
CI I ' N~IJ.CH,
o N~cH, methylbenzamide


~ ~ NHi


CH,


2g ., H~ 565.1N-(3-aminopropyl)-N-[1-[7-chloro-3-(3-


~ methylbenzyl)-4-oxo-3,4-


~N O
c~ ~ ' N~N~H, dihydroquinazolin-2-yl]-2-(dimethylamino)-


O NCH, 2-oxoethyl]-4-methylbenzamide


~ ~ NHz


CH,


-45-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Com oundStructure MH+ Name


30 OxF 631.1 -(3-aminopropyl)-N-[1-{7-chloro-4-oxo-3-
N


I F [ 4-(trifluoromethoxy)benzyl]-3,4-
~


~ d ihydroquinazolin-2-yl)-2-(dimethylamino)-
w N
O


I 2 -oxoethyl]-4-methylbenzamide
~~~"~



~ I NH,


CFh


31 ~ 581.5 N-(3-aminopropyl)-N-[1-[7-chloro-3-(4-


~ ~ c hlorobenzyl)-4-oxo-3,4-


N d ihydroquinazolin-2-yf]-2-(dimethylamino)-
~
H~


' 2 -oxoethyl]-4-methylbenzamide
o~~H,


w I NH,


CHI


3~ ~~ ~ 616.0 N-(3-aminopropyf)-N-[1-[7-chloro-3-(2,5-


dichlorobenzyl)-4-oxo-3,4-


of I ' N~N'CH' dihydroquinazolin-2-yl]-2-(dimethylamino)-


O NCH, 2-oxoethyl]-4-methylbenzamide
~


l
~ NHz


CH,


33 ~ ; 616.0 6-
N-(3-aminopropyl)-N-[1-[7-chloro-3-(2


o' ~~ :
dichlorobenzyl)-4-oxo-3,4-


ci I ' N N NCH~ dihydroquinazolin-2-yl]-2-(dimethylam~no)-
~


2-oxoethyl]-4-methylbenzamide
~H,
~


I
Hz


CH,


34 i ~ o,~H, 577.1 N-(3-aminopropyl)-N-[1-[7-chloro-3-(3-


methoxybenzyl)-4-oxo-3,4-


cI I ' N N N'CH' dihydroquinazolin-2-yl]-2-(dimethylamino)-


2-oxoethyl]-4-methylbenzamide


0
~ ~ Hz


CH3


35 ~ I 597.1 N-(3-aminopropyl)-N-[1-[7-chloro-3-(2-



naphthylmethyl)-4-oxo-3,4-


N dihydroquinazolin-2-yl]-2-(dimethylamino)-


N N.CFh
NCH, 2-oxoethyl]-4-methylbenzamide


y ~ NHz


CH,


-46-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Com ~ Structure MH+ Name
ound


36 F F 615.1 N-(3-aminopropyl)-N-[1-{7-chloro-4-oxo-3-


o ~ ~ F [3-(trifluoromethyl)benzyl]-3,4-


i ; N ~ ~ dihydroquinazolin-2-yl}-2-(dimethylamino)-
H,


~~H, 2-oxoethyl]-4-methylbenzamide
~


I
Hz


CH,


37 o F 631.1 N-(3-aminopropyl)-N-[1-{7-chloro-4-oxo-3-
~


[3-(trifluoromethoxy)benzyl]-3,4-
' F dihydroquinazolin-2-yl}-2-(dimethylamino)-
N
I


' N
N'CH
CI
~
3


-oxoethyl]-4-methylbenzamide
~H,


I ~ Hz


CH3


38 F F F F 683.1 N-(3-aminopropyl)-N-[1-{3-[3,5-


bis(trifluoromethyl)benzyl]-7-chloro-4-oxo-


" ,~H, 3,4-dihydroquinazolin-2-yl}-2-
I
~


o (dimethylamino)-2-oxoethyl]-4-
~H,


methylbenzamide
~


w
Hz


CH,


3g I ~ off 563.1 N-(3-aminopropyl)-N-[1-(7-chloro-3-(3-


~ hydroxybenzyl)-4-oxo-3,4-
li
l
2
di
th
l
i
dih
d
i
2


CI ' N~N'CH'
nazo
n-
]-
-(
me
y
am
no)-
y
roqu
-y


o 'N~~", 2-oxoethyl]-4-methylbenzamide


~ ~ NHz


CH3


40 H, i ~ H, 575.1 N-(3-aminopropyl)-N-[1-[7-chloro-3-(3,5-


~ dimethylbenzyl)-4-oxo-3,4-


CI ' N~N'CH'
dihydroquinazolin-2-yl]-2-(dimethylamino)-


o N~H, 2-oxoethyl]-4-methylbenzamide


~ NHz


CH,


41 \ i 623.2 N-(3-aminopropyl)-N-[1-[3-(1,1'-biphenyl-



3-yimethyl)-7-chloro-4-oxo-3,4-


N dihydroquinazolin-2-yl]-2-(dimethylamino)-


a ~ N~N'CH~
O NCH, 2-oxoethyl]-4-methylbenzamide


~ ~ NHz


CH,


-47-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Com oundStructure MN+ Name


42 ~ ,N 572.1 N-(3-aminopropyl)-N-[1-[7-chloro-3-(3-
~


cyanobenzyl)-4-oxo-3,4-dihydroquinazolin-
'


~I ~ ~ N N N.CH 2-yl]-2-(dimethylamino)-2-oxoethyl]-4-
~
3


~H, methylbenzamide


~ ~ Hz


CH,


43 . 592.1 N-(3_aminopropyl)-N-[1-[7-chloro-3-(3-
N,


~
O nitrobenzyl)-4-oxo-3,4-dihydroquinazolin-
~ ~


2-yl]-2-(dimethyfamino)-2-oxoethyl]-4-
i .CH
~


~~H, methylbenzamide


~ ~ NHz


CH,


44 irN\ 548.1 N-(3-aminopropyl)-N-[1-[7-chloro-4-oxo-3-


~~ (pyridin-4-ylmethyl)-3,4-dihydroquinazolin-
N
I


~ 2-yl]-2-(dimethylamino)-2-oxoethyl]-4-
~
' N~N-CH~
CI


N1 CH, methylbenzamide
~


w
NHz
CHI


45 ~\ ~I 616.0 N-(3-aminopropyl)-N-[1-[7-chloro-3-(3,4-


O ~
dichlorobenzyl)-4-oxo-3,4-


N' O
~i dihydroquinazolin-2-yl]-2-(d~methylammo)-
'OH'
~~


N 2-oxoethyl]-4-methylbenzamide
CI
O N' CH,
y


'li
W I NHz


CH,


46 i ~ 561.1 N-(3-aminopropyl)-N-{1-(3-benzyl-7-
~


chloro-4-oxo-3,4-dihydroquinazolin-2-yl)-2-
I [ethyl(methyl)amino]-2-oxoethyl}-4-


J~CH
CI
' N~


I methylbenzamide
,
o N\ oH,
l~
~ NHz


CH,


47 H, i ~ C~, 589.1 N-(3-aminopropyl)-N-{1-[7-chloro-3-(3,5-


dimethylbenzyl)-4-oxo-3,4-
N O


dihydroquinazolin-2-yl]-2-
cl ' ~
~H


~H [ethyl(methyl)amino]-2-oxoethyl}-4-
,


methylbenzamide
~
' i


W
Hz


CH,


-48-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Com oundStructure MH+ Name


48 -H, 581.1 -(3-aminopropyl)-N-{1-[7-chloro-3-(3-
I N


~
' methoxybenzyl)-4-oxo-3,4-


I ~ N O
o. ~ N~N~CH d ihydroquinazolin-2-yl]-2-


, [ methyl)amino]-2-oxoethyl)-4-
N ~~ ( ethyl


~ methylbenzamide


NH,


cH,


4g F i ~ F 583.0 N-(3-aminopropyl)-N-[1-[7-chloro-3-(3,5-


' d ifluorobenzyl)-4-oxo-3,4-
O


N-CH, d ihydroquinazolin-2-yl]-2-(dimethylamino)-
C~ I ' N N
~


~H3 -oxoethyl]-4-methylbenzamide


w ~ NHi


CH,


50 % ~ 582.5 N-(3-aminopropyl)-N-[1-{7-chloro-3-[(6-


N chloropyridin-2-yl)methyl]-4-oxo-3,4-


~ ~ ~ ~~N,CH, dihydroquinazolin-2-yl)-2-(dimethylamino)-


O N' CH, -oxoethyl]-4-methylbenzamide


w ~ NHi


CH,


51 ~ 582.5 N-(3-aminopropyl)-N-[1-{7-chloro-3-[(2-


~ chloropyridin-4-yl)methyl]-4-oxo-3,4-
' droquinazolin-2-yl)-2-(dimethylamino)-
~ dih


' N~N-~", y
~


O NCH, 2-oxoethyl]-4-methylbenzamide


~ ~ NHi


CH,


52 ~ i 598.1 N-(3-aminopropyl)-N-[1-[7-chloro-4-oxo-3-


o i N (quinolin-2-ylmethyl)-3,4-


N o dihydroquinazolin-2-yl]-2-(dimethylamino)-


I
~~","' 2-oxoethyl]-4-methylbenzamide
~


w I
Hz


CH,


Using the procedure described in Example 4, the above compounds were shown
to have an KSP inhibitory activity at an IC~p of less than about 50 pM.
Certain of the
compounds have an ICSp of less than about 1 ~,M, and certain others of the
compounds
have an ICsp of less than about 100 nM.
-49-



CA 02546932 2006-05-23
WO 2005/051922 PCT/US2004/039448
Example 4
Assay for Determining KSP Activity
In this example, a representative in vitro assay for determining KSP activity
is
described.
Purified microtubules from bovine brain were purchased from Cytoslceleton Inc.
The motor domain of human KSP (EgS, KNSLl) was cloned and purified to a purity
of
greater than 95%. Biomol Green was purchased from Affinity Research Products
Ltd.
Microtubules and the KSP motor protein were diluted in assay buffer (20 mM
Tris-HCI, pH 7.5, 1 mM MgCl2, 10 mM DTT and 0.25 mg/ml BSA) to a concentration
of
35 ug/ml for microtubules and 45 nM for KSP. The microtubulelKSP mixture was
then
pre-incubated at 37°C for 10 min to promote the binding of KSP to,
microtubules.
ATP was also diluted to a concentration of 300 uM in the same assay buffer. To
each
well of the testing plate (384 well plate) containing 1.25 uL of compounds in
DMSO or
DMSO only, 25 uL of ATP solution. To start the reaction, 25 uL of
microtubulelKSP
solution was added to the ATP/compound mixture. The plates were incubated at
room
temperature for 1 hr. At the end of incubation period, 65 uL of Biomol Green
was added
to each well. The plates were incubated for 5-10 min and then the absorbance
at 630 nm
was determined. Biomol Green reagent is a malachite green based dye that
detects the
release of inorganic phosphate. Developed color signal was read using a Victor
II
reader. The concentration of each compound for 50% inhibition (ICSO) was
calculated by
nonlinear regression using either XLFit for Excel or Prism data analysis
software by
GraphPad Software Inc.
While the preferred embodiment of the invention has been illustrated and
described, it will be appreciated that various changes can be made therein
without
departing from the spirit and scope of the invention.
-50-

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2004-11-24
(87) PCT Publication Date 2005-06-09
(85) National Entry 2006-05-23
Dead Application 2010-11-24

Abandonment History

Abandonment Date Reason Reinstatement Date
2009-11-24 FAILURE TO PAY APPLICATION MAINTENANCE FEE
2009-11-24 FAILURE TO REQUEST EXAMINATION

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2006-05-23
Maintenance Fee - Application - New Act 2 2006-11-24 $100.00 2006-05-23
Registration of a document - section 124 $100.00 2006-05-29
Maintenance Fee - Application - New Act 3 2007-11-26 $100.00 2007-10-16
Registration of a document - section 124 $100.00 2008-09-02
Maintenance Fee - Application - New Act 4 2008-11-24 $100.00 2008-10-21
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
NOVARTIS VACCINES AND DIAGNOSTICS, INC.
Past Owners on Record
CHIRON CORPORATION
CONSTANTINE, RYAN N.
DESAI, MANOJ C.
LAGNITON, LIANA M.
WANG, WEIBO
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2006-05-23 1 62
Claims 2006-05-23 9 357
Description 2006-05-23 50 2,417
Representative Drawing 2006-08-07 1 4
Cover Page 2006-08-08 1 36
Correspondence 2008-12-03 2 51
Assignment 2006-05-23 3 98
PCT 2006-05-23 4 169
Assignment 2006-05-29 10 344
Prosecution-Amendment 2006-10-16 11 333
Assignment 2008-09-02 10 327